US6248712B1 - Urokinase-type plasminogen activator receptor - Google Patents

Urokinase-type plasminogen activator receptor Download PDF

Info

Publication number
US6248712B1
US6248712B1 US08/442,108 US44210895A US6248712B1 US 6248712 B1 US6248712 B1 US 6248712B1 US 44210895 A US44210895 A US 44210895A US 6248712 B1 US6248712 B1 US 6248712B1
Authority
US
United States
Prior art keywords
par
binding
plasminogen
pro
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US08/442,108
Other languages
English (en)
Inventor
Keld Danø
Francesco Blasi
Ann Louring Roldan
Maria Vittoria Cubellis
Maria Teresa Masucci
Ettore Appella
W.D. Schleuning
Niels Behrendt
Ebbe Rønne
Peter Kristensen
Jari Pöllänen
Eeva-Marjatta Salonen
Ross W. Stephens
Hannele Tapiovaara
Antti Vaheri
Lisbeth Birk Møller
Vincent Ellis
Leif Røge Lund
Michael Ploug
Charles Pyke
Lászl{acute over (o )} Patthy
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cancerforskningsfondet af 1989
Original Assignee
Cancerforskningsfondet af 1989
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/319,052 external-priority patent/US5891664A/en
Application filed by Cancerforskningsfondet af 1989 filed Critical Cancerforskningsfondet af 1989
Priority to US08/442,108 priority Critical patent/US6248712B1/en
Application granted granted Critical
Publication of US6248712B1 publication Critical patent/US6248712B1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6456Plasminogen activators
    • C12N9/6462Plasminogen activators u-Plasminogen activator (3.4.21.73), i.e. urokinase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1808Epidermal growth factor [EGF] urogastrone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/49Urokinase; Tissue plasminogen activator
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21073Serine endopeptidases (3.4.21) u-Plasminogen activator (3.4.21.73), i.e. urokinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57446Specifically defined cancers of stomach or intestine
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/86Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving blood coagulating time or factors, or their receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/972Plasminogen activators
    • G01N2333/9723Urokinase

Definitions

  • the present invention relates to a method for preventing or counteracting localized proteolytic activity in a mammal, in particular a human, the method comprising inhibiting the activation of plasminogen to plasmin by preventing the binding of a receptor binding form of urokinase-type plasminogen activator (in the following termed u-PA) to a u-PA receptor in the mammal and thereby preventing the u-PA from converting plasminogen into plasmin; the invention also relates to a pure u-PA receptor (in the following termed u-PAR), to DNA coding for the u-PAR, to the production of u-PAR or parts thereof for use as a therapeutic or diagnostic component, to u-PAR antibodies and the production of u-PA receptor binding u-PA molecules for use as a therapeutic or diagnostic component.
  • u-PA urokinase-type plasminogen activator
  • the invention relates to the regulation of the activity of a receptor binding form of u-PA, the activation of pro-u-PA to u-PA by plasmin and the regulation of the number of u-PARs on the cell and the binding affinity of the u-PAR/u-PA binding as well as the therapeutic aspects of these findings.
  • the invention relates to the detection of u-PAR by labelled u-PA.
  • urokinase-type plasminogen activator (u-PA) has been found in all mammalian species so far investigated.
  • u-PA urokinase-type plasminogen activator
  • u-PA plays a role in the degradative phase of inflammation, and there have also been reports that u-PA interferes with the lymphocyte-mediated cytotoxicity against a variety of cells, and a direct role of u-PA in the cytotoxic effect of natural killer cells has been proposed.
  • a role of u-PA has been proposed in angiogenesis and in endothelial cell migration, a process important in tumor growth.
  • u-PA is produced by many cultured cell types of neoplastic origin. It has been found that explants of tumor tissue released more u-PA than the corresponding normal tissue. u-PA has been identified in extracts from human lung, colon, endometrial, breast, prostate and renal carcinomas, human melanomas, murine mammary tumors, the murine Lewis lung tumor, and in ascites from human peritoneal carcinomatosis. An immunohistochemical study of invasively growing and metastasing Lewis lung carcinomas in mice consistently showed the presence of u-PA, but also a pronounced heterogenecity in the content of u-PA in different parts of the individual tumors. A high u-PA content was found in areas with invasive growth and degradation of surrounding normal tissue, while other areas were devoid of detectable u-PA. The u-PA was located in the cytoplasm of the tumor cells and extracellularly surrounding the tumor cells.
  • Degradation of the surrounding normal tissue is a central feature of invasiveness of malignant tumors.
  • the constant finding of u-PA in malignant tumors and the findings indicating that u-PA plays a role in tissue degradation in normal physiological events have led to the assumption that u-PA plays a similar role in cancer development.
  • the hypothesis of u-PA playing a role in tissue destruction involves the assumption that plasmin, together with other proteolytic enzymes, degrades the extracellular matrix. It is noteworthy in this context that most components of the extracellular matrix can be degraded by plasmin. These include laminin, fibronectin, proteoglycans, and possibly some types of collagen, but not all.
  • plasmin can activate latent collagenases which in turn can degrade the other types of collagen (see Dan ⁇ et al., 1988).
  • the extracellular matrix is composed of glycoproteins such as fibronectin and laminin, collagen and proteoglycans. Extracellular matrix becomes focally permeable to cell movement only during tissue healing and remodelling, inflammation, and neoplasia.
  • Liotta (1986) has proposed a three-step hypothesis: The first step is tumor cell attachment via cell surface receptors. The anchored tumor cell next secretes hydrolytic enzymes (or induces host cells to secrete enzymes) which can degrade the matrix locally (including degradation of the attachment components).
  • Matrix lysis most probably takes place in a highly localized region close to the tumor cell surface.
  • the third step is tumor cell locomotion into the region of the matrix modified by proteolysis.
  • invasion of the matrix is not merely due to passive growth pressure but requires active biochemical mechanisms.
  • a requirement for the regulation of a proteolytic cascade system in extracellular processes is the precise localization of its initiation and progression.
  • cellular receptors for various components are known and serve to localize reactions that either promote or terminate the reaction sequence (Müller-Eberhard, 1988, Mann et al., 1988).
  • t-PA tissue-type plasminogen activator
  • u-PA is located at the membrane of the tumor cells (Skriver et al., 1984), and recent findings indicate that at cell surfaces, u-PA is generally bound to a specific receptor and that this localization may be crucial for the regulation of u-PA catalyzed plasminogen activation in time and space (see Blasi et al., 1987).
  • u-PAR The cellular receptor for u-PA (u-PAR) was originally identified in blood monocytes and in the monocyte-like U937 cell line (Vassalli et al., 1985), and its presence has been demonstrated on a variety of cultured cells, including several types of malignant cells (Stoppelli et al., 1985, Vassalli et al., 1985, Plow et al., 1986, Boyd et al., 1988a, Nielsen et al., 1988), human fibroblasts (Bajpai and Baker, 1985), and also in human breast carcinoma tissue (Needham et al., 1987).
  • the receptor binds active 54 kD u-PA, its one-polypeptide chain proenzyme, pro-u-PA (see below), as well as 54 kD u-PA inhibited by the active site reagent DFP, but shows no binding of the low molecular weight (33 kD) form of active u-PA (Vassalli et al., 1985; Cubellis et al., 1986).
  • binding to the receptor does not require the catalytic site of u-PA, and in agreement with these findings, the binding determinant of u-PA has been identified in the amino-terminal part of the enzyme, in a region which in the primary structure is remote from the catalytic site.
  • the receptor binding domain is located in the 15 kD amino-terminal fragment (ATF, residues 1-135) of the u-PA molecule, more precisely within the cysteine-rich region termed the growth factor region as this region shows homologies to the part of epidermal growth factor (EGF) which is responsible for binding to the EGF receptor.
  • the amino acid residues which appear to be critical for binding are located within the sequence 12-32 (numbered 1-21 in SEQUENCE ID NO:32) of u-PA. (Appella et al., 1987). Synthetic peptides have been constructed that inhibit the binding of very low (100 nM) concentrations. The lack of cross-reactivity between the murine and the human peptides indicates that the binding between u-PA and u-PAR is strongly species specific.
  • Binding of u-PA to u-PAR is specific in the sense that as yet no other protein has been found to compete for binding to the receptor, though several proteins structurally related to u-PA, including t-PA and plasminogen, have been tested (Stoppelli et al., 1985, Vassalli et al., 1985, Nielsen et al., 1988). Fragments of u-PA containing only the receptor binding domain, e.g.
  • ATF ensure specificity of the binding to the receptor, since other molecules that might bind u-PA (protease nexin and the specific plasminogen activator inhibitors PAI-1 and PAI-2) recognize the catalytically active region (Stoppelli et al., 1985; Nielsen et al., 1988). PAI-1 is able to form a covalent complex with u-PA but not with pro-u-PA (Andreasen et al., 1986).
  • the number of receptors can be regulated by the addition of various agents such as phorbol myristate acetate (PMA) in U937 cells (Stoppelli et al., 1985, Nielsen et al., 1988), epidermal growth factor in A431 cells (Blasi et al., 1986) and HeLa cells (Estreicher et al., 1989) and dimethylformamide in colon carcinoma cells (Boyd et al., 1988b).
  • PMA phorbol myristate acetate
  • the purified u-PAR preparation shows essentially one radiolabelled band after SDS-PAGE followed by autoradiography. This analysis, however, does not show the purity of the preparation as it does not detect unlabelled proteins that may be present in an amount that may be higher than that of the u-PAR. Similar considerations hold true for a recent study by Estreicher et al. (1989), in which attempts at purifying u-PAR were done from cells that had been surface-labelled with 125 I.
  • u-PA is released from many types of cultured cells as a single-chain proenzyme with little or no plasminogen activating capacity (Nielsen et al., 1982, Skriver et al., 1982, Eaton et al., 1984, Kasai et al., 1985, Pannell and Gurewich 1987).
  • this proenzyme can be converted to its active two-chain counterpart.
  • Such assays for pro-u-PA are self-activating and are strongly influenced by small amounts of contaminating or generated two-chain u-PA or plasmin. As discussed in detail elsewhere (Petersen et al., 1988), it is therefore possible that the high activity of one-chain u-PA found in these studies was apparent and not due to intrinsic activity of single-chain u-PA. Consistent with this interpretation is a report on a variant of recombinant single-chain u-PA which by site-directed mutagenesis was made partly resistant to plasmin cleavage. This variant of single-chain u-PA had an activity that in coupled assays was 200-fold lower than that of two-chain u-PA (Nelles et al., 1987).
  • pro-u-PA is the predominant form of u-PA in intracellular stores, and it also constitutes a sizable fraction of the u-PA in extracellular fluids (Skriver et al., 1984, Kielberg et al., 1985). Extracellular activation of pro-u-PA may therefore be a crucial step in the physiological regulation of the u-PA pathway of plasminogen activation.
  • the plasmin-catalyzed activation of pro-u-PA provides a positive feedback mechanism that accelerates and amplifies the effect of activation of a small amount of pro-u-PA.
  • the initiation of the u-PA pathway of plasminogen activation under physiological conditions involves triggering factors that activate pro-u-PA as described herein.
  • Mutants of human single-chain pro-u-PA in which lysine 158 is changed to another amino acid are not, or are only to a small extent, converted to active two-chain u-PA (Nelles et al., 1987).
  • u-PA has been found to be unevenly distributed, distinctly located at cell-cell contact sites and at focal contacts that are the sites of closest apposition between the cells and the substratum (Pöllänen et al., 1987, 1988, Hébert and Baker 1988). u-PA was not detected in the two other types of cell-substratum contact, i.e. close contacts and fibronexuses, making it an intrinsic component at focal contact sites (Pöllänen et al., 1988). u-PA at the focal contact sites is receptor-bound (Hébert and Baker, 1988).
  • the focal contact sites are located at the termini of actin-containing microfilament bundles, the so-called stress fibers or actin cables (Burridge, 1986). These sites contain several structural components (actin, talin) and regulatory factors (the tyrosine kinase protooncogene products P60 src , P120 gag-abl , P90 gag-yes , P80 gag-yes ), that are all located on the cytoplasmic side (see Burridge, 1986).
  • Plasminogen as well as plasmin, binds to many types of cultured cells, including thrombocytes, endothelial cells and several cell types of neoplastic origin (Miles and Plow, 1985, Hajjar et al., 1986, Plow et al., 1986, Miles and Plow 1987, Burtin and Fondaneche, 1988).
  • the binding is saturable with a rather low affinity for plasminogen (K D 1 ⁇ M).
  • binding of plasmin appears to utilize the same site as plasminogen, but the binding parameters for plasmin indicate that more than one type of binding site for plasminogen and plasmin may exist.
  • plasmin and plasminogen bind with almost equal affinity (Plow et al., 1986), while on others plasmin apparently binds with a higher affinity (K D 50 nM) than plasminogen (Burtin and Fondaneche, 1988).
  • the binding is inhibited by low amounts of lysine and lysine analogues and appears to involve the kringle structure of the heavy chains of plasminogen and plasmin (Miles et al., 1988).
  • the binding capacity varies between cell types and in many cell types is quite high (10 5 -10 7 binding sites per cell). The chemical nature of the binding sites are not known.
  • a membrane protein, GPIIb/IIIa seems to be involved in the binding of plasminogen to thrombocytes (Miles et al., 1986) and, particularly on thrombin-stimulated thrombocytes, also fibrin may be involved in plasminogen binding (Miles et al., 1986).
  • the thrombocyte protein thrombospondin forms complexes (K D 35 nM) with plasminogen (Silverstein et al., 1984).
  • immobilized laminin (Salonen et al., 1984) and fibronectin (Salonen et al., 1985) bind plasminogen (K D 3 nM and 90 nM, respectively)
  • Receptor-bound pro-u-PA can be activated by plasmin (Cubellis et al., 1986) and, at least in part, receptor-bound two-chain u-PA retains its ability to activate plasminogen (Vassalli et al., 1985).
  • Ellis et al. (1989) recently published evidence indicating that the reactions leading to plasminogen activation can take place when single-chain u-PA and plasminogen are added to U937 cells, and that they occur more efficiently when both plasminogen and pro-u-PA are bound to the surface.
  • This experiment was performed in the absence of serum, i.e. under conditions where the plasminogen activation with the preparations used by Ellis et al. will also take place in solution (cf. Ellis et al., 1987), and these studies do not exclude the possibility that one or more of the processes involved (e.g. the plasminogen activation catalyzed by two-chain u-PA) actually occurred when the u-PA was not receptor-bound.
  • the present invention is based upon the discovery that under conditions similar to those present extracellularly in the intact organism (i.e. in the presence of serum containing inhibitors of plasmin and of plasminogen activators), plasminogen activation initiated by endogenous u-PA occurs only when the u-PA is receptor-bound, upon the provision of pure u-PA receptor, and upon the provision of the possibility of producing the u-PA receptor or characteristic and valuable sequences thereof or analogues to sequences thereof by recombinant DNA technology.
  • new and potentially extremely valuable therapeutic, prophylactic and diagnostic methods and products, together with associated basic methods and products are provided by the present invention.
  • Plasminogen binds to cell surfaces, and surprisingly it was found that a large part, if not all, of the cell surface plasminogen activation is catalyzed by surface-bound u-PA, and that binding of plasmin to the surface is necessary for the activation of pro-u-PA.
  • most of the cell surface u-PA is present in its single-chain proenzyme form (pro-u-PA), while addition of plasminogen leads to the formation of receptor-bound two-chain u-PA. The latter reaction is catalyzed by cell-bound plasmin.
  • Receptor-bound u-PA is accessible to inhibition by endogenous PAI-1 and by added PAI-2, while the cell-bound plasmin is inaccessible to serum inhibitors.
  • a model for cell-surface plasminogen activation can be made in which plasminogen binding to cells is followed by plasminogen activation by trace amounts of bound active u-PA, to form bound plasmin, which in turn serves to produce more active u-PA from bound pro-u-PA.
  • This exponential process is subject to regulation by endogenous PAI-1, and limited to the pericellular space.
  • the new findings include the requirement, in the presence of serum, for binding of plasminogen, the ability of bound u-PA under these conditions to activate plasminogen, the presence of pro-u-PA on the cells, the ability of bound plasmin to activate pro-u-PA, and the ability of endogenous plasminogen activator inhibitor PAI-1, as well as added plasminogen activator inhibitor PAI-2, to regulate the surface plasminogen activation.
  • tumor cells can acquire the broad-spectrum proteolytic activity of plasmin, bound to their surface in such a way that it is protected from inactivation by serum protease inhibitors, and ideally situated to be employed in the degradation of the pericellular matrix.
  • Binding of u-PA to its receptor localizes u-PA not only to the cell surface, but focalizes it to distinct parts of the surface that at least in some cell types are the cell-cell and cell-substrate contact.
  • the location of pro-u-PA at the focal contact sites suggests that u-PA catalyzed plasminogen activation is involved in the breakdown of the contacts, e.g. during cell movement.
  • a selective activation of pro-u-PA at these sites provides a means of obtaining a directional pericellular proteolysis.
  • pro-u-PA activation might be intracellularly initiated and mediated by a transmembrane signal through the u-PA receptor.
  • Human tumor cells are very commonly found to secrete plasminogen activator of the urokinase type (u-PA). By this means they are able to recruit the proteolytic potential available in the high concentration of plasminogen in plasma and other body fluids.
  • the invasive properties of tumor cells may be at least partly dependent on their proteolytic capability mediated through the broad spectrum of activity of plasmin and including its indirect actions in activating other latent proteases, such as collagenases.
  • the expression of protease activity by tumor cells facilitates their penetration of basement membranes, capillary walls and interstitial connective tissues, allowing spread to other sites and establishment of metastases.
  • a stepwise pathway of pericellular proteolysis geared to cell migration can be envisaged: binding of u-PA and plasminogen to the cell surface will lead to extracellular proteolysis and to the local severing of cell-cell and cell-substrate connections. This region of the cell is therefore free to move and this will transpose u-PA to a region in which PAI-1 is present. PAI-1 will inactivate u-PA and in the absence of local proteolytic activity, the cell will form new connections with the matrix, a process required for further migration.
  • the expression of the u-PA gene is finely regulated by a variety of agents that affect cell growth; however, until recently very little was known of the regulation of the u-PAR function and synthesis. It is known that the affinity of the u-PA receptor can be modified by e.g. the tumor promoter PMA. This indicates that the cells are endowed with mechanisms that modulate the u-PA:u-PAR interaction. While this interaction appears to act at the level of the receptor itself, the effect of the plasminogen activator inhibitors demonstrates a second level of modulation, i.e. at the level of the active ligand itself. It is possible that the two levels of regulation might actually be interconnected, i.e. that the binding of the inhibitor to surface-bound u-PA influences the affinity of the receptor.
  • the change in affinity is a regulatory mechanism capable of modifying the ratio between soluble and surface-bound u-PA, i.e. regulating the location of u-PA. It is possible that the effects on synthesis and affinity of u-PAR normally take place either in different cells or in the same cells, but in response to different stimuli.
  • the physiological signal for the affinity-regulating mechanism may be connected with the level and possibly the fine localization of the u-PA activity on the cell surface.
  • Example 8 show that blocking binding to the u-PA receptor, or to PAI-1 and/or PAI-2 inhibitors, should result in an increase in the half life of therapeutically administered pro-u-PA and u-PA, thus allowing a decrease of the therapeutically efficient dosage.
  • Example 1 Further characterization of the interaction of u-PA and u-PAR required the purification of the u-PAR.
  • the number of u-PAR produced by the monocyte-like cell U937 can be increased several fold by phorbol esters like PMA. This fact was used to produce sufficient quantities of the receptor for purification.
  • Example 1 a complete purification of the u-PA receptor is described, involving temperature-induced phase separation of a detergent extract from cells, and affinity chromatography with immobilized DFP-inactivated u-PA. This resulted in a preparation that shows one band at approximately 55-60 kD after SDS-PAGE and silver staining, with a load of approximately 1 ⁇ g of the receptor.
  • the purified protein could be chemically cross-linked with u-PA. Its amino acid composition and N-terminal sequence were determined (30 residues, some of which with some uncertainty). It was found to be heavily N-glycosylated, deglycosylation resulting in a protein with an apparent molecular weight of about 30-35 kD. The apparent molecular weight of u-PAR from different cell lines and from PMA-stimulated and non-stimulated U937 cells varied somewhat. This heterogeneity disappeared after deglycosylation and was thus due to differences in glycosylation of u-PAR from the various sources.
  • Example 1 The modulation of the u-PAR molecules demonstrated in Example 1 may represent an important feature in the regulation of extracellular proteolysis and thus in the degradation of the extracellular matrix and basement membrane components, processes that are at the core of cell migration and invasiveness.
  • Example 2 isolation of a ligand binding domain of u-PAR is identified and characterized. This provides potentially therapeutically valuable information on peptides that may inhibit the ligand binding.
  • u-PAR is produced as a 313 residues long protein with a 282 residues long hydrophilic N terminal part (probably extracellular) followed by 21 rather hydrophobic amino acids (probably a trans-membrane domain).
  • the potential extracellular part is organised in 3 repeats with striking homologies, particularly with respect to the pattern of cysteines. This may indicate the presence of distinct domains that may bind different ligands.
  • the receptor purification and cDNA cloning allowed to recognize that the u-PAR is at least in some cases terminally processed and anchored to the cell surface via a glycolipid anchor, and that the surface location can be regulated by the phospholipase PI-PLC, but not by the phospholipases A 2 and D (Example 4). Furthermore, it was found that also harvest fluid from cells that were not treated contain some free u-PAR indicating release from the cells that may be mediated by an endogenous phospholipase. This may be a physiological mechanism and it is possible that measurement of free receptor, e.g. in serum, may be a diagnostically valuable indicator of some pathological processes.
  • the u-PAR cDNA was used to show that u-PAR mRNA could be regulated in some cell types by substances such as PMA, dexamethasone, mEGF and TGF- ⁇ -1 (Example 5).
  • substances such as PMA, dexamethasone, mEGF and TGF- ⁇ -1 (Example 5).
  • the findings indicate that these and similar substances may be therapeutically useful in regulating u-PAR synthesis.
  • u-PAR cDNA was used to produce fragments of u-PAR antisense mRNA which proved useful for the detection of u-PAR mRNA in tissue sections by in situ hybridization. Particularly interesting is the finding that u-PAR mRNA is consistently present in human colon carcinomas and is located in cells at the invasive front of the tumors, thus indicating the production of u-PAR by these cells and a role of u-PAR in localizing u-PA at these sites.
  • Example 7 it is demonstrated that after incubation of monolayer cultures of human HT-1080 fibrosarcoma cells with purified native human plasminogen in serum containing medium, bound plasmin activity can be eluted from the cells with tranexamic acid, an analogue of lysine.
  • the bound plasmin is the result of plasminogen activation on the cell surface; plasmin activity is not taken up onto cells after deliberate addition of plasmin to the serum containing medium.
  • the cell surface plasmin formation is inhibited by an anti-catalytic monoclonal antibody to u-PA, indicating that this enzyme is responsible for the activation.
  • Plasmin released from the cells was partially inactivated in the serum medium. As long as the plasmin remained bound, it was protected from serum inhibitors but could be inhibited by aprotinin or an anti-catalytic monoclonal antibody.
  • Plasmin has been shown to be produced in this condition, yet one would expect that it would be inactivated by serum inhibitors. If a significant fraction is bound to cells, however, this may escape inhibiton and retard development of healing tissue, until an effective inhibitor is applied externally.
  • Example 7 it is shown that preincubation of the cells with DFP-incubated u-PA led to a decrease in surface-bound plasmin, indicating that a large part, if not all, of the cell surface plasminogen activation was catalyzed by surface-bound u-PA.
  • plasminogen activator activity is largely dependent on addition of exogenous u-PA.
  • u-PA is administered in a binding step followed by washing of the cells and assay.
  • the activity can be competed by receptor binding u-PA antagonists, e.g. synthetic peptides and DFP-treated u-PA, and can be inhibited by the addition of PAI-1.
  • PAI-1 also binds to, and inhibits the activity of receptor-bound u-PA in U937 cells.
  • Example 8 also shows that PAI-1/u-PA complexes bind the receptor of U937 cells with the same specificity and affinity as free u-PA.
  • PAI-1 is able to interact also with u-PA pre-bound on the receptor on the U937 cells. This results in the formation of a typical covalent PAI-1/u-PA complex that is not detectably internalized, and in the inhibiton of the u-PA activity.
  • the affinity of complexed u-PA is slightly decreased compared to free u-PA. Possibly the presence of the bulky PAI-1 molecule may pose a problem of steric hindrance.
  • Example 8 it is further shown that when u-PA/PAI-1 complexes are bound to the U937 cells, they are subsequently degraded and internalized.
  • Example 9 shows that PAI-1 and PAI-2 rather rapidly inhibit receptor-bound u-PA, although the respective association rate constants are lower than those for inhibition of u-PA in solution.
  • u-PA binding of u-PA to solubilized (Example 10) or purified (Example 13) u-PAR inhibits the ability of u-PA to activate plasminogen in solution, in contrast to the stimulation of the activity which is observed when u-PA is bound to u-PAR on a cell surface concomitantly with surface binding of plasminogen.
  • Purified u-PAR also inhibits plasmin catalyzed pro-u-PA activation in solution (Example 13).
  • Polyclonal antibodies were developed by immunization of mice with purified u-PAR (Example 11). These antibodies precipitated 125 I-labelled purified u-PAR in a dose-dependent manner, with a significant precipitation being obtained by the antiserum in a dilution of 1:7500.
  • the antiserum was found to immunocapture radiolabelled u-PAR, and in an ELISA immobilized u-PAR in an amount of 1 ng was detected with the immune serum diluted 1:8000.
  • Western blotting the antibodies detected both purified u-PAR and u-PAR in the crude detergent phase of extracts of PMA-treated U937 cells.
  • the Example also includes a description of methods for development of monoclonal antibodies using the above-mentioned methods for screening of hybridomas for antibody production.
  • Example 11 describes that antibodies to u-PAR can be used to specifically prevent ligand binding. It is furthermore shown (Example 10) that u-PAR antibodies inhibit u-PA catalyzed cell surface plasminogen activation. These or similar antibodies may also be used to specifically target bacterial or vegetable toxins with the purpose of destroying potentially metastatic tumor cells.
  • Example 12 a method for the visualization of the u-PAR on cells and in tissue sections is described, using biotinylated DFP-treated u-PA followed by incubation with streptavidin-fluorescin isothiocyanate.
  • the method is very sensitive, and its specificity can readily be tested by competition experiments (e.g. with the amino-terminal fragment of u-PA (ATF), t-PA, EGF, etc.).
  • the present invention provides inhibition of receptor binding of u-PA as a means of inhibiting some of its physiological functions in relationship to therapeutic prevention of localized proteolytic activity, e.g. invasion and metastasis of cancer cells, inflammatory bowel disease, premalignant colonic adenomas, septic arthritis, osteoarthritis, rheumatoid arthritis (for which a direct involvement of excess u-PA production has been demonstrated), osteoporosis, cholesteatoma, and a number of skin and corneal diseases for which an excess plasminogen activation has been shown to be the pathogenetic cause, such as corneal ulcers, keratitis, epidermolysis bullosa, psoriasis, and pemphigus.
  • localized proteolytic activity e.g. invasion and metastasis of cancer cells, inflammatory bowel disease, premalignant colonic adenomas, septic arthritis, osteoarthritis, rheumatoid arthritis (for which a direct involvement
  • u-PA receptors are present in several blood and endothelial cells, their regulation might also significantly affect intravascular fibrinolytic activity in physiological, pathological and pharmacological conditions.
  • the above-mentioned diseases would be the first obvious targets for a therapy based on administration of substances that block or decrease cell surface plasminogen activation.
  • a contraceptive effect is expected of measures that inhibit receptor binding.
  • the therapy and prophylaxis will involve systemic or topical treatment with agents that block or reduce receptor bound plasminogen activator activity, such as will be explained below.
  • the present invention also provides valuable reagents and methods for diagnostic or research purposes, such as the u-PA receptor (u-PAR), the u-PAR cDNA, anti u-PAR antibodies, and u-PA antagonists like modified u-PA and pro-u-PA molecules, obtained by chemical, biological, or synthetic means, such as explained below.
  • u-PAR u-PA receptor
  • cDNA u-PA receptor
  • anti u-PAR antibodies anti u-PAR antibodies
  • u-PA antagonists like modified u-PA and pro-u-PA molecules, obtained by chemical, biological, or synthetic means, such as explained below.
  • u-PA urokinase type plasminogen activator
  • t-PA tissue-type plasminogen activator
  • Both u-PA and t-PA are widely used in thromboembolic therapy.
  • the identification of the u-PA/t-PA receptors and of agents that prevent binding and/or internalization and degradation of u-PA/t-PA to these receptors can be exploited to increase the half life of the administered substance by administering the substance together with an agent which will prevent or reduce binding of the substance to its receptor in the cardiovascular system and therefore to reduce the doses and their side effects.
  • the invention relates to a method for preventing or counteracting localized proteolytic activity in a mammal, in particular a human, comprising inhibiting the activation of plasminogen to plasmin by preventing the binding or inducing the specific degradation of a receptor binding form of u-PA to a u-PA receptor (u-PAR) in the mammal and thereby preventing u-PA from converting plasminogen into plasmin.
  • u-PAR u-PA receptor
  • the term “localized proteolytic activity” is intended to designate a proteolytic activity which is located at one or several distinct regions in a human body, or at distinct cells, as opposed to an overall proteolytic activity exerting itself substantially anywhere in the body.
  • the localized proteolytic activity can be inhibited generally in a mammal, in particular a human, or locally.
  • the term “preventing or counteracting” is intended to designate a situation where the binding of u-PA to u-PAR is completely inhibited, or a situation where the binding is sufficiently inhibited so as to inhibit the undesired effect of the plasminogen activator.
  • the term “inducing the specific degradation” is intended to designate a process by which the receptor-binding form of u-PA is degraded in a specific manner, e.g. internalized such as described in Example 8 in which the specific degradation is induced by adding PAI-1.
  • a receptor binding form of u-PA is intended to mean any form of u-PA possessing a site that binds to a site at a u-PAR, that is to say that the u-PA contains the u-PAR binding site.
  • the receptor binding form of u-PA can thus be pro-u-PA, u-PA, an amino-terminal fragment of u-PA, a u-PA that is irreversibly inhibited by e.g. diisopropyl fluorophosphate (DFP), p-nitrophenyl-p′-guanidinobenzoate (NPGB), or any other inhibitor or any other modification of u-PA that can bind to a u-PAR.
  • DFP diisopropyl fluorophosphate
  • NPGB p-nitrophenyl-p′-guanidinobenzoate
  • a u-PAR indicates that even though the polypeptide part of u-PAR in a species might be the same for all u-PARs, there is a plurality of u-PARs as for example the carbohydrate part or the mechanism of surface attachment of the u-PAR can be different. It may even be so that some cells, e.g. cancer cells, have substantially different u-PARs which might have important therapeutic significance as it might be possible to block the binding of u-PA to u-PARs residing on a cancer cell without affecting the binding of u-PA to u-PARs on non-pathological cells or of specifically killing cancer cells that express u-PAR.
  • u-PA urokinase-type plasminogen activator
  • plasminogen By cleavage at Arg 560 , plasminogen is activated to the broad spectrum protease plasmin.
  • preventing u-PA from converting plasminogen into plasmin is therefore meant that this activation by u-PA is substantially inhibited or a situation where the activation is sufficiently inhibited so as to inhibit or reduce the undesired effect of the plasmin.
  • the prevention of the binding of a receptor binding form of u-PA to a u-PAR is, e.g. suitably performed by blocking the u-PAR by administration, to the mammal, of a substance binding to the u-PAR so as to occupy a site of the receptor to which a receptor binding form of u-PA is normally bound, the substance being administered in an amount effective to reduce the binding of the receptor binding form of u-PA to the receptor.
  • blocking the u-PAR means that a substance that is not able to activate plasminogen to plasmin is bound to u-PAR, preferably by a substantially irreversible binding, thereby preventing a receptor binding form of u-PA of catalyzing the conversion of plasminogen into plasmin.
  • binding to a u-PAR so as to occupy a site of the receptor to which a receptor binding form of u-PA is normally bound is intended to mean that the substance binds to the u-PAR so that a receptor binding form of u-PA can not be bound to the u-PAR.
  • the prevention of the binding of a receptor binding form of u-PA to a u-PAR may be performed by administering a modification of u-PA which has retained its capability of binding to the u-PAR, but which is not capable of converting plasminogen to plasmin, to the mammal.
  • a modification of u-PA is u-PA inhibited at its catalytically active site with a substantially irreversible inhibitor, i.e. a substance which binds to the catalytically active site by a substantially irreversible bond.
  • a substantially irreversible inhibitor i.e. a substance which binds to the catalytically active site by a substantially irreversible bond.
  • DFP-u-PA diisopropyl fluorophosphate
  • the modification of u-PA may also be obtained by binding an antibody to its catalytically active site region.
  • the antibody may be either polyclonal antibodies or monoclonal antibodies and may be prepared as described in greater detail below.
  • u-PA amino-terminal fragment of u-PA (ATF-u-PA) (cf. Stoppelli et al., 1985).
  • the prevention of the binding of a receptor binding form of u-PA to u-PAR may also be performed by administering a substance comprising a sequence which is identical or substantially identical to a site of u-PA which binds to the u-PAR; such substance may for example be a molecule comprising a sequence which is identical or substantially identical to a u-PAR binding site of u-PA amino residues 12-32, which are known to be involved in the receptor binding of u-PA.
  • Another way of preventing the binding of a receptor binding form of u-PA to a u-PAR is to administer a u-PAR or a u-PA-binding modification thereof to the mammal so as to occupy the cell receptor-binding site of u-PA, thereby preventing the receptor binding form of u-PA from binding to the cell-bound receptor.
  • a complete u-PAR but rather a water-soluble form thereof, in other words a part thereof comprising a u-PA binding sequence.
  • u-PAR can be solubilized by removal of the glycerol-phosphoinositol anchor, e.g. with a lipase as shown in Example 4.
  • u-PAR plasminogen activator inhibitors
  • PAI-1 plasminogen activator inhibitor Type 1
  • PAI-2 plasminogen activator inhibitor Type 2
  • the prevention of the binding of a receptor binding form of u-PA to a u-PAR may also be obtained by administering a modification of pro-u-PA which has retained its capability of binding to the u-PAR, but which is not capable of being converted into u-PA.
  • a modification of pro-u-PA is one in which the sequence of u-PA normally cleavable by plasmin has been changed so that the u-PA is not cleaved by plasmin.
  • An example of this is pro-u-PA in which Lys 158 has been substituted with Glu or Gly by site-directed mutagenesis.
  • a very interesting method of preventing the binding of a receptor binding form of u-PA to a u-PAR and thereby preventing the cell surface plasminogen activation is the use of antibodies against u-PAR such as demonstrated in Example 11 (prevention of binding) and Example 10 (prevention of cell surface plasminogen activation).
  • the antibodies may be polyclonal antibodies, preferably of high specificity such as the antibodies illustrated in Example 11, or a monoclonal antibody.
  • the antibody may be an antibody that is reactive with non-carbohydrate moieties of the u-PAR, or it may be an antibody that is reactive with carbohydrate moieties of the u-PAR, the latter permitting a valuable distinction between target cells where cells expressing distinct variants of u-PAR are the cells involved in the undesired proteolysis.
  • the antibodies may be administered in various ways as described below.
  • Another strategy for preventing or counteracting localized proteolytic activity in a mammal, in particular a human comprises inhibiting the activation of plasminogen to plasmin by substantially reducing the activity of a receptor-bound form of u-PA by administering, to the mammal, a plasminogen activator inhibitor in a sufficient amount to inhibit the activation of plasminogen.
  • the plasminogen activator inhibitor may be PAI-1 or PAI-2 which, according to the present invention, have been found to inhibit u-PA, also when it is receptor-bound.
  • Another strategy for preventing or counteracting localized proteolytic activity in a mammal, in particular a human comprises inducing the selective internalization of the receptor-bound u-PA by, for example, blocking its activity by administering the specific inhibitor PAI-1 or increasing PAI-1 synthesis with hormones (estrogens, glucocorticoids, polypeptide hormones), cytokines (interleukins, interferons, TNF) or growth factors (EGF, IGF-1, IGF-2, PDGF, FGF, TGF ⁇ , TGF ⁇ ) and any other factor that induces PAI-1 synthesis, thereby causing u-PA degradation and internalization.
  • hormones estrogens, glucocorticoids, polypeptide hormones
  • cytokines interleukins, interferons, TNF
  • growth factors EGF, IGF-1, IGF-2, PDGF, FGF, TGF ⁇ , TGF ⁇
  • Another strategy for inducing intracellular u-PA degradation consists in administering compounds that would induce dimerization of receptors, such as PAI-1 dimers, as in other receptors dimerization appears to be involved in internalization.
  • Another strategy for preventing or counteracting localized proteolytic activity is removal of u-PAR from cell surfaces by treatment with an agent which destroys the glycerol-phosphoinositol anchor, e.g. a phosholipase, such as PI-PLC as described in Example 4.
  • an agent which destroys the glycerol-phosphoinositol anchor e.g. a phosholipase, such as PI-PLC as described in Example 4.
  • the agent will preferably be administered locally.
  • Yet another strategy for preventing or counteracting localized proteolytic activity in a mammal comprises inhibiting the activation of plasminogen to plasmin by altering the binding affinity of the u-PA/u-PAR by modifying u-PAR in the mammal (e.g. by treatment with the phorbol ester PMA or with EGF) and thereby preventing u-PA from converting plasminogen into plasmin.
  • a reduction of the binding affinity may be obtained by administering a substance selected from the group consisting of hormones, growth factors (such as epidermal growth factor [EGF]) or cytokines.
  • the PAI-1 induced internalization of receptor-bound u-PA can also be exploited to selectively kill tumor cells by administering a PAI-1 derivative which is covalently bound (by chemical or genetic methods) to a bacterial or plant toxin.
  • a PAI-1 derivative which is covalently bound (by chemical or genetic methods) to a bacterial or plant toxin.
  • the cells Upon internalization of the u-PA-PAI-1-toxin complex, the cells can be selectively killed by the action of the toxin.
  • u-PA u-PA
  • a therapeutic effect may be obtained by administering u-PAR or a derivative thereof having u-PAR function or by conferring to the cells of the patient the ability to produce functionally intact u-PAR or a derivative thereof having u-PAR function by transfection with the u-PAR cDNA or a variant thereof.
  • the synthesis of u-PAR may be increased by administration of various hormones, growth factors, or cytokines, e.g. dexamethazone, mEGF, and TGF- ⁇ -1 as indicated by the findings described in Example 5.
  • a completely different, and potentially therapeutically very valuable application of u-PAR, solubilized u-PAR and variants thereof is as an inhibitor of u-PA-catalyzed plasminogen activation in solution.
  • the hitherto known specific inhibitors of plasminogen activators that is, PAI-1 and PAI-2, inhibit both u-PA and t-PA.
  • Purified u-PAR as well as u-PAR solubilized by removal of the glycerol-phosphoinositol anchor inhibits u-PA in solution as demonstrated in Examples 10 and 13.
  • u-PAR does not bind t-PA.
  • u-PAR is contemplated to be more advantageous than PAI-1 and PAI-2 in cases where specific inhibition of u-PA is needed.
  • Potentially very valuable is also the therapeutic use of u-PAR, solubilized u-PAR and variants thereof for inhibiting the activation of the virtually inactive single-chain pro-u-PA molecule to active two chain u-PA.
  • u-PAR extracellular part of u-PAR consists of three repeats with considerable mutual homologies (Example 3) renders it probable that it can bind different ligands, that is, that it can bind other ligands apart from the proven binding of u-PA. It would be justified to assume that some of these may involve yet unknown plasminogen receptors or plasminogen binding sites because of the strong enhancing effect obtained by concomitant binding of u-PA and plasminogen to the cell surface as described in Example 7.
  • Potential alternative ligands for u-PAR may also be proteins located at cell-cell and focal cell-substratum contact sites because of the preferential location of receptor-bound u-PA at these sites in some cell types.
  • Variants or parts of u-PAR that inhibit binding of such ligands may be valuable in inhibition of cell surface plasminogen activation, and prevention of binding of u-PAR to such ligands may be functionally important and therapeutically valuable in a broad spectrum of diseases.
  • u-PAR exists in various forms, such as the glycosylation variants described in Example 1, the variants with different sensitivity to the lipase PI-PLC suggested by the findings described in Example 4, and the variants in ability to stimulate cell surface plasminogen activation found on PMA-stimulated and PMA-non-stimulated U937 cell, respectively, as described in Example 10.
  • some of these forms may be preferentially changed. Therapy directed against correcting some distinct forms may therefore be particularly therapeutically valuable in such diseases.
  • the administration of the various above-mentioned principles to a mammal, preferably a human being, may be performed by any administration method which is suitable for administering proteins or peptides or antibodies.
  • Typical administration routes are parenteral, oral, nasal, topical or rectal administration.
  • the active ingredient to be administered should be formulated in a manner which will protect the active ingredient against degradation, in particular by enzymes.
  • the parenteral administration is the safest way of administering proteins and peptides.
  • the parenteral administration route should be selected dependent on where the active ingredient is to be released, e.g. intravenously, intramuscularly or subcutaneously, etc. It is also important to consider the necessity of “packing” the active ingredient in a suitable manner in order to
  • suitable measures should be taken to protect the active ingredient from enzymatic degradation in the gastrointestinal tract, e.g. by packing the active ingredient in such a way that it will not be released from the formulation (i.e. the pharmaceutical composition) until it has reached the site where either the active ingredient is to exert its activity locally (i.e. in the gastrointestinal tract) or from where the absorption may take place (e.g. M-cells in the colon).
  • Nasal administration is an administration form which is presently intensively investigated in order to provide absorption of substances of the peptide type from the nasal cavity. In principle, this may take place in two ways, firstly by using enhancers, and secondly by using the bioadhesion principle in which the active ingredient may be maintained for a long period of time at a suitable domain in the nose.
  • Topical administration may be performed by formulating the active ingredient in a salve, an ointment, a lotion, a creme, etc.
  • compositions of the invention may for example include pharmaceutically acceptable excipients adapted to the character of the active ingredients in accordance with the above discussion. Suitable excipients may include liposomes and/or microspheres.
  • the preparation of the pharmaceutical compositions may be performed in accordance with methods described in the literature for compositions of the types described herein.
  • a suitable pericellular (extracellular) concentration thereof is in the range of from 1 ⁇ g/ml to well above 10 ⁇ g/ml, such as, e.g., 100 ⁇ g/ml, but it is also noted from FIG. 11 that even concentrations smaller than 1 ⁇ g/ml do have a significant effect for which reason a practical lower limit could be set at 0.1 ⁇ g/ml.
  • the treatments will normally be continued for weeks or often months and are suitably combined with treatment with other medicaments against the conditions in question.
  • Another strategy of treating the conditions and diseases mentioned above is to target a cell that contains a u-PAR on the surface by a medicament, comprising administering the medicament bound to a substance that binds to a u-PAR.
  • the substance may be a receptor binding form of u-PA, or it may be an antibody against u-PAR such as a polyclonal or a monoclonal antibody, e.g. an antibody particularly directed to a variant of u-PAR present in a cancer cell type.
  • the medicament may typically be an anti-cancer agent such as an alkylating agent, e.g. melphalan, chlorambucil, busulfan, cisplatin, thiotepa, an antimetabolite such as methotrexate, fluracil, azathioprin, an antimitoticum, typically vincristine, vinblastine, or an antibiotic such as doxorubicin, daunorubicin or bleomycin.
  • the medicament may also comprise bacterial or other toxins.
  • Another important aspect of the present invention is a method of targeting a cell that contains a u-PAR on the surface by a diagnostic, comprising administering the diagnostic bound to a receptor binding form of u-PA.
  • the diagnostic may be a radioactive substance which is physiologically tolerable such as, e.g., technetium.
  • An important field of the present invention is a number of diagnostic methods which methods, or the importance thereof, are based upon the findings according to the invention.
  • One important aspect thereof is a method of detecting a u-PAR in a tissue section comprising treating the tissue section with a substance that binds to a u-PAR, and visualizing the presence of the bound substance.
  • the substance may in principle be any of the above substances which bind to u-PAR, but it is especially preferred that the substance is an antibody, in particular a labelled antibody or an unlabelled antibody which is subsequently detected by an immunostaining method.
  • the antibody may be a polyclonal or monoclonal antibody, and particularly interesting antibodies are antibodies that distinguish between various forms of u-PAR. A detailed description of diagnostic kits, materials and methods based on antibodies is given further below.
  • the substance which binds to u-PAR may also be a form of u-PA, either labelled, for example biotinylated DFP-treated u-PA that subsequently is detected by streptavidin-fluorescin isothiocyanate, or an unlabelled form which is subsequently detected by immunostaining.
  • Another field of the invention is the use of antibodies against a u-PAR for the quantification of the u-PAR in biological material using antibodies aganist the u-PAR. While this method may be performed using either polyclonal or monoclonal antibodies, an interesting embodiment uses a combination of polyclonal and monoclonal antibodies, the monoclonal antibodies being more specific and the polyclonal antibodies generally having a higher binding affinity.
  • the quantification method may be of the ELISA type or may be a radioimmunoassay. These assays may be produced by methods known per se.
  • One aspect of the invention relates to a method of producing pure u-PAR, the method comprising subjecting a u-PAR-containing material to affinity chromatography with immobilized antibodies to u-PAR and eluting the u-PAR, e.g. under acidic conditions.
  • the present invention also relates to pure u-PAR.
  • pure u-PAR has been made for the first time in accordance with the present invention.
  • Pure u-PAR in glycosylated form shows, in an SDS-PAGE at a load of approximately 1 ⁇ g, substantially one and only one silver stained band having an apparent molecular weight in the range of about 55-60 kD.
  • the presence of substantially one and only one silver stained band in this SDS-PAGE is a proof of the purity of the u-PAR.
  • Another proof of the purity of the u-PAR is the presence of a single amino-terminal amino acid sequence in purified u-PAR preparations.
  • the glycosylated u-PARs upon deglycosylation, were all found to have an identical electrophoretic mobility (corresponding to substantially one and only one band at about 30-35 kD in an SDS-PAGE), indicating that the peptide part of the molecule is identical in all cases.
  • pure u-PAR in glycosylated form may be prepared from a biological material containing u-PAR by temperature-induced phase separation of detergent extracts followed by affinity chromatography purification with immobilized DFP-u-PA.
  • the detergent is preferably a non-ionic detergent such as a polyethylene glycol ether, e.g. Triton X-114.
  • the temperature was found to be relatively critical in the range of 34-40° C., such as about 37° C., for 10 minutes.
  • the pure u-PAR in unglycosylated form may be prepared by deglycosylation with, e.g., peptide/N-glycosidase F.
  • the present invention also relates to a novel method of purification of u-PAR, exploiting the ability of phospholipase C to release the receptor in the medium, thereby providing a direct method of preparing and purifying an extracellular, soluble form of u-PAR which is able to bind u-PA and which can be used as a u-PA scavenger.
  • a 24-mer nucleotide probe was synthesized and used to screen a library to identify and isolate recombinant clones carrying the cDNA for u-PAR.
  • the identity of the cDNA clones was confirmed by comparing the nucleotide sequence of this cDNA clone with the amino terminal sequence of the purified u-PAR, and by expressing said cDNA in mouse L cells and assaying their u-PA-binding properties.
  • One aspect of the invention relates to a polypeptide comprising a characteristic amino acid sequence derived from a u-PAR which polypeptide comprises at least 5 amino acids and up to the complete sequence of u-PAR as shown below as the DNA sequence (SEQ ID NO:22) and the deducted amino acid sequence (SEQ ID NO:23) of the clone p-uPAR-1.
  • the signal peptide is underlined and the first 30 amino acids, the sequence of which has been determined on the purified protein with an Applied Biosystems gas phase sequencer (see Example 1), are overlined.
  • the putative transmembrane domain is doubly underlined.
  • the star symbols indicate the potential N-linked glycosylation sites.
  • the invention relates to any polypeptide comprising at least 5 amino acids and up to the complete sequence of u-PAR from amino acid 1 to 313, and any analogue to such a polypeptide.
  • the polypeptide may be coupled to a carbohydrate or lipid moiety. It may typically be glycosylated as mentioned above.
  • the term “characteristic amino acid sequence derived from the u-PAR” is intended to mean an amino acid sequence, such as an epitope, which comprises amino acids constituting a substantially consecutive stretch (in terms of linear or spatial conformation) in u-PAR, or amino acids found in a more or less non-consecutive conformation in u-PAR, which amino acids constitute a secondary or tertiary conformation having interesting and useful properties, e.g. as therapeutics or diagnostics.
  • amino acids present at different positions in u-PAR but held together e.g. by chemical or physical bonds, e.g. by disulphide bridges, and thereby forming interesting tertiary configurations are to be understood as “characteristic amino acid sequences”.
  • the characteristic amino acid sequence may comprise a consecutive subsequence of the amino acid sequence of u-PAR of greater or smaller length or a combination of two or more parts of such subsequences which may be separated by one or more amino acid sequences not related to u-PAR.
  • the characteristic amino acid sequences may be directly bonded to each other.
  • epitope refers to a sequence or subsequence of the polypeptides of the invention or a derivative or an analogue thereof capable of stimulating or interacting with immunocompetent cells, especially epitopes against which antibodies showing desirable properties in regard to diagnosis can be raised.
  • analogue is used in the present context to indicate a protein or polypeptide of a similar amino acid composition or sequence as the characteristic amino acid sequence derived from the u-PAR, allowing for minor variations which do not have an adverse effect on the immunogenicity of the analogue.
  • the analogous polypeptide or protein may be derived from mammals or may be partially or completely of synthetic origin.
  • the present invention also relates to a substantially pure polypeptide which is recognized by an antibody raised against or reactive with a polypeptide comprising the amino acid sequence defined above.
  • the term “substantially pure” is understood to mean that the polypeptide in question is substantially free from other components, e.g. other polypeptides or carbohydrates, which may result from the production and/or recovery of the polypeptide or otherwise be found together with the polypeptide.
  • the high purity of the polypeptide of the invention is advantageous when the polypeptide is to be used for, e.g., the production of antibodies.
  • the substantially pure polypeptide may be used in a lower amount than a polypeptide of a conventional lower purity for most purposes.
  • the purification of the polypeptide of the invention may be performed by methods known to a person skilled in the art, but particularly the low concentrations of u-PAR in biological material and the strongly hydrophobic nature of the receptor has hitherto hampered its purification. Now, however, the combination of temperature-induced phase separation of detergent extracts of cells and affinity chromatography with immobilized DFP-treated u-PA has led to its successful purification in amounts high enough (100-200 ⁇ g) to have enabled a partial amino acid sequencing and further characterization.
  • the invention in another aspect, relates to a DNA fragment comprising a nucleotide sequence encoding the u-PAR described above.
  • the DNA fragment may be used in a method of preparing the u-PAR or parts thereof by recombinant DNA techniques or as a diagnostic agent (i.e. a DNA probe).
  • a diagnostic agent i.e. a DNA probe.
  • the use of the DNA fragment of the invention in the production of a recombinant polypeptide e.g. by inserting the fragment in a suitable vector, transforming the vector into a suitable host organism (microorganism or cultured animal cell), cultivating the organism so as to produce the polypeptide and subsequently recovering the polypeptide from the organisms) includes a number of advantages.
  • the DNA fragment of the invention may also be used as a diagnostic agent for the detection of mRNA encoding u-PAR or parts thereof in a sample, which diagnostic agent comprises a labelled DNA sequence which is homologous with a DNA sequence coding for at least part of u-PAR.
  • the pure u-PAR (natural or recombinant) of the invention may be used in the preparation of polyclonal or monoclonal antibodies.
  • the antibodies may be used for the identification and/or quantification of at least part of the above described polypeptide present in a sample thus making it possible to diagnose diseases related to presence of abnormal numbers of the u-PAR on the surface of mammalian cells.
  • the sample may be any part of the human organism, e.g. be a body fluid or tissue part containing the polypeptide, e.g. a tissue sample such as a biopsy, e.g.
  • a bone marrow tissue sample a blood sample, a urine sample, a sample of cerebrospinal fluid, serum, plasma or any product prepared from blood or lymph, secretions or any sample obtained from a human cavity containing cells with a u-PA receptor.
  • the polypetide of the invention may be coupled to a carbohydrate, a lipid moiety or modified in other ways, e.g. phosphorylated or hydroxylated.
  • the polypeptide may be glycosylated, the coupled carbohydrate moiety having molecular weights of 20-30 kD in the natural molecule.
  • Coupling of the polypeptide to one or more moieties may for instance be due to a posttranslational modification of the polypeptide performed by an organism producing the polypeptide or a modification resulting from chemical synthesis.
  • the polypeptide of the invention may also be a fusion protein in which characteristic amino acid sequence(s) from u-PAR is/are fused to another polypeptide sequence.
  • the polypeptide to which the characteristic amino acid sequence(s) from u-PAR is/are fused may be one which results in an increased expression of the protein when expressed in an organism, or facilitates or improves the purification and recovery of the fusion protein from said organism in terms of a more easy and economical recovery, or confers to the u-PAR the property of inhibiting u-PA (as it would be in the case of a u-PAR-PAI-1 fusion).
  • the characteristic amino acid sequence(s) from u-PAR is/are preferably fused to a polypeptide sequence which may be specifically recognized by a cleaving agent, e.g. a chemical such as cyanogen bromide, hydroxylamine and 2-nitro-5-thiocyanobenzoate, or an enzyme, e.g. a peptidase, proteinase or protease, e.g. trypsin, chlostripain, and staphylococcal protease or factor Xa.
  • a cleaving agent e.g. a chemical such as cyanogen bromide, hydroxylamine and 2-nitro-5-thiocyanobenzoate
  • an enzyme e.g. a peptidase, proteinase or protease, e.g. trypsin, chlostripain, and staphylococcal protease or factor Xa.
  • one aspect of the present invention relates to a DNA fragment encoding the polypeptide of the invention.
  • the invention relates to a DNA fragment comprising substantially the nucleotide sequence (1), or a subsequence thereof coding for a subsequence of the polypeptide of the invention.
  • A represents adenine
  • T represents thymidine
  • G represents guanine
  • C represents cytosine
  • This nucleotide sequence encodes the entire protein part of u-PAR.
  • the DNA sequence shown above has been established as described in Example 3.
  • the cDNA of the u-PAR represents a rather rare clone, based on the fact that it is expressed at the most at 800,000 molecules/cell. It has in fact been found with a frequency of less than 6 ⁇ 10 ⁇ 6 .
  • the cDNA is about 1.4 kb long based on its restriction map (FIG. 4 B), has a 5′ untranslated sequence of about 40 residues and an about 40 nucleotides long poly-A stretch at the 3′ end.
  • DNA hybridization is a useful method. Hybridization may be performed as follows: Pure DNA comprising the gene encoding u-PAR from the plasmid p-uPAR-1 is prepared using the large scale method described in Maniatis et al. (1982), pages 86-96. More specifically, the u-PAR gene may be excised from the plasmid by digestion of the plasmid DNA with suitable restriction enzymes. The insert is then separated from the plasmid DNA by use of agarose gel electrophoresis. The insert is labelled by any labelling principle, such as the ones disclosed herein.
  • the foreign DNA to be examined is coupled to a matrix, e.g. a nitrocellulose filter.
  • the filter is subjected to a suitable treatment suited to the kind of matrix employed so as to couple the DNA to the matrix, in the case of a nitrocellulose filter e.g. by baking the filter at a temperature of 80° C. for 2 hours.
  • the membrane is exposed to a prehybridization solution of a composition, at a temperature and for a period of time recommended suited to the membrane in question.
  • the membrane is then placed in the hybridization solution containing the labelled denatured DNA probe obtained from the p-uPAR-1 plasmid (the u-PAR gene). Hybridization is preferably carried out overnight at a suitable temperature.
  • the membrane is then washed and incubated with a volume of 50 ml 2 ⁇ SSC at 65° C. for 30 minutes. The procedure is repeated once.
  • the membrane is then incubated in 15 ml 2 ⁇ SSC containing 0.1% SDS. Incubation is performed at 65° C. for 30 minutes. All incubations including prehybridization and washings are performed with gentle agitation.
  • the filter is air-dried and wrapped in a suitable plastic wrap (e.g. Saran Wrap), the filter is then applied to an x-ray film so as to obtain an auto-radiographic image. Exposition is preferably carried out at ⁇ 70° C. with intensifying screens for a period of time which is determined by the positive control used.
  • any hybridization of the foreign DNA and the u-PAR gene is an indication of similarity of the two DNA probes, i.e. that the foreign DNA is a DNA fragment of the invention.
  • Another approach of determining similarity between DNA sequences is by determining the nucleotide sequence of the DNA sequence to be compared with the DNA sequence of the invention by conventional DNA sequencing analysis, and comparing the degree of homology with the DNA sequence of the invention.
  • a degree of homology of at least about 70%, e.g. at least about 80% such as at least about 95% is obtained.
  • the DNA fragment of the invention may comprise a nucleotide sequence encoding a polypeptide fused in frame to the nucleotide sequence encoding the characteristic amino acid sequence with the purpose of producing a fused polypeptide.
  • the fused sequence may be inserted into an appropriate vector which is transformed into a suitable host organism.
  • the DNA fragment of the invention may be inserted in the vector in frame with a gene carried by the vector, which gene encodes a suitable polypeptide.
  • the host organism which might be of eukaryotic or prokaryotic origin, for instance a yeast or a mammalian cell line, is grown under conditions ensuring expression of the fused sequence after which the fused polypeptide may be recovered from the culture by physico-chemical procedures, and the fused polypeptide may be subjected to gel filtration and affinity chromatography using an antibody directed against the antigenic part(s) of the fused polypeptide.
  • the polypeptide of the invention and the polypeptide to which it is fused may be separated, for instance by suitable proteolytic cleavage, and the polypeptide of the invention may be recovered, e.g. by affinity purification or another suitable method.
  • the DNA fragment may also comprise a suitable nucleotide sequence controlling the expression of the DNA fragment.
  • the regulatory nucleotide sequence is conveniently a part of the expression vector used for the production of the polypeptides, when such a vector is employed.
  • the DNA fragment described above may be obtained directly from genomic DNA or by isolating mRNA and transferring it into the corresponding DNA sequence by using reverse transcriptase producing cDNA.
  • obtaining the DNA fragment from genomic DNA it is derived directly by screening for genomic sequences, hybridizing to a DNA probe prepared on the basis of the full or partial amino acid sequence of u-PAR.
  • the DNA is of complementary DNA (cDNA) origin, it may be obtained by preparing a cDNA library on the basis of mRNA from cells containing a u-PAR or parts thereof. Hybridization experiments may then be carried out using synthetic oligonucleotides as probes to identify the cDNA sequence encoding the u-PAR or part thereof.
  • cDNA differs from genomic DNA in, e.g. that it lacks certain transcriptional control elements and introns which are non-coding sequences within the coding DNA sequence. These elements and introns are normally contained in the genomic DNA.
  • the DNA fragment may also be of synthetic origin, i.e. prepared by conventional DNA synthesizing method, e.g. by using a nucleotide synthesizer. The DNA fragment may also be produced using a combination of these methods.
  • DNA or RNA fragment comprising the sequence complementary to the above DNA sequence or a part thereof or the mRNA corresponding to said DNA sequence. It is contemplated that a DNA or RNA fragment complementary to at least part of the mRNA corresponding to the polypeptide of the invention is effective in arresting the translation of the polypeptide in the human cells, and thereby inhibiting the synthesis of u-PAR polypeptides. In other systems, it has been shown that DNA or RNA fragments complementary to the mRNA encoding a given protein is capable of arresting the translation of the protein.
  • an antisense-oncogene in a human cell line as described by Holt, J. T. et al., Proc. Natl.
  • antisense DNA or RNA complementary to virus mRNA has been shown to be able to inhibit the infection rate of the SP-phage in an E. coli strain (as described in Hirashima, A. et al., Proc. Natl. Acad. Sci. USA, 1986, 83, 7726-7730), or of the HIV-virus in an infected human CD4 cell line (Reitz, M. et al., Proc. Natl. Acad. sci., 1987, 84, 7706-7710).
  • the above DNA or RNA fragment should comprise a number of nucleotides which is sufficient for obtaining the desired specificity and hybridization of the DNA or RNA fragment to the mRNA corresponding to the polypeptide of the invention.
  • the DNA or RNA fragment is of a size which allows the safe transport of the fragment through the cell membrane, i.e. without any substantial disruption of the fragment transported over the membrane.
  • the DNA or RNA fragment should have a size of at least about 5 nucleotides, preferably at least about 8 nucleotides.
  • the DNA or RNA fragment should have a size of at the most about 100 nucleotides, preferably at the most about 80 nucleotides.
  • the DNA or RNA fragment having a size of about 10-60 nucleotides, such as about 12-50 nucleotides, e.g. about 14-40 nucleotides, preferably about 15-25 or 15-22 nucleotides is useful.
  • the DNA or RNA fragment may be complementary to any part of the mRNA, e.g. to a part of the mRNA comprising the ribosomal binding site or part thereof, or the start codon for the gene encoding the polypeptide of the invention, or to a sequence which is repeated one or more times.
  • the DNA or RNA fragment may comprise multiple phosphate-modified oligodeoxyribonucleotides such as oligo-alkyl phosphotriesters, oligomethylphosphonates or oligophosphorothioates to improve the resistance against nucleases or the transport across cell membranes.
  • the DNA or RNA fragment may be prepared by conventional methods, e.g. the methods outlined above.
  • the invention relates to an expression vector which is capable of replicating in a host organism and which carries a DNA fragment as described above.
  • the vector may be any vector which conveniently can be subjected to recombinant DNA procedures, the choice of vector often depending on the host cell into which it is to be introduced.
  • the vector may either be one which is capable of autonomous replication, i.e. a vector which exists as an extrachromosomal entity, the replication of which is independent of chromosomal replication, such as a plasmid, or a vector which is replicated together with the host chromosome, such as a bacteriophage.
  • plasmids such as natural or synthetic plasmids, eg. plasmids related to pBR322 such as pEX 1-3, the pRIT-family, the pUC-family and the like, and viruses such as adenovirus, vaccinia virus, retrovirus, Baculo virus, Epstein-Barr-virus, SV40-related virus and bovine papilloma virus.
  • suitable bacteriophages include M13 and lambda.
  • the invention also relates to an organism which carries and is capable of expressing a DNA fragment as defined above and which not in its native form expresses said DNA fragment.
  • the DNA fragment may be carried on a vector as described above or may be integrated in the genome of the organism.
  • suitable organisms include microorganisms such as bacteria, yeasts, fungi and higher eucaryotic organisms or cells including plant and mammalian cells.
  • higher organisms such as animals, e.g. sheep, cattle, goats, pigs, etc. is contemplated to be useful as host organisms for the production of the polypeptide of the invention.
  • the present invention also relates to a method of producing the polypeptides described above.
  • the polypeptides are prepared using recombinant DNA-technology e.g. the methods disclosed in Maniatis et al. op. cit. More specifically, the polypeptides may be produced by a method which comprises cultivating or breeding an organism carrying a DNA-fragment encoding a characteristic amino acid sequence from an u-PAR, e.g. the above described DNA fragment, under conditions leading to expression of said DNA fragment, and subsequently recovering the polypeptide from the organism.
  • the organism which is used for the production of the polypeptide may be a higher organism, e.g. an animal, or a lower organism, e.g. a microorganism.
  • the DNA fragment encoding the characteristic amino acid sequence from an u-PAR should be introduced in the organism.
  • the DNA fragment is inserted in an expression vector, e.g. a vector as defined above, which is subsequently introduced into the host organism.
  • the DNA fragment may also be directly inserted in the genome of the host organism.
  • the insertion of the DNA fragment in the genome may be accomplished by use of a DNA fragment as such or cloned in bacteria, phage lambda or other vectors, carrying the DNA fragment and being capable of mediating the insertion into the host organism genome.
  • the insertion of the DNA fragment into an expression vector or into the genome of the host organism may be accomplished as described e.g. by Colbere-Garapin F. et al., J. Molec. Biol., 150; 1-14 (1981): A New Dominant Hybrid Selective Marker for Higher Eucaryotic Cells.
  • a higher organism e.g. an animal
  • transgenic techniques known in the art may be employed for the production of the polypeptide. Examples of suitable animals are sheep, cattle, pigs, etc.
  • the DNA encoding the polypeptide of the invention is suitably inserted into the genome of the animal in such a position that the polypeptide of the invention is expressed together with a polypeptide which inherently is expressed by the animal, preferably a polypeptide which is easily recovered from the animal, e.g. a polypeptide which is secreted by the animal, e.g. a milk protein, or the like.
  • the DNA fragment of the invention is inserted in the genome of the animal in frame with the DNA fragment encoding the polypeptide inherent to the animal so as to obtain expression of a fusion protein comprising on the one hand the polypeptide of the invention and on the other hand the polypeptide related to the host organism, e.g. the animal.
  • the resulting fusion protein may then be subjected to posttranslational modification so as to obtain the polypeptide of the invention.
  • the DNA fragment when using an expression vector for the production of the polypeptide of the invention, may be inserted in frame with a second DNA fragment encoding another polypeptide so as to obtain an expression of fusion protein.
  • the polypeptide of the invention comprises one or more distinct parts, e.g. being a fusion protein comprising on the one hand characteristic amino acid sequence(s) from u-PAR and on the other hand amino acid sequence(s) constituting a polypeptide which is not related to u-PAR
  • the DNA fragments encoding each of these polypeptides may be inserted in the genome or expression vector separately or may be coupled before insertion into the genome or expression vector by use of conventional DNA techniques such as described in Maniatis et al. op. cit.
  • the conditions under which the organism producing the polypeptide of the invention is cultured or breeded should of course be adapted to the organism employed.
  • Conventional cultivation and breeding techniques may be employed.
  • the cultivation is e.g. carried out in a culture medium conventionally used for fermentation purposes, e.g. Luria Broth medium, and under conditions with respect to pH, temperature, aeration, etc. suited to the type of microorganism in question, e.g. as disclosed in Maniatis et al. op. cit.
  • the polypeptide is recovered or isolated from the organism.
  • the polypeptide may be isolated or recovered from the culture by a method comprising one or more affinity chromatography and/or size chromatography steps, and optionally employing a step using an antibody reactive with and/or being raised against said polypeptide.
  • the procedure used for recovering of the polypeptide depends on the kind of host organism used as well as the polypeptide produced.
  • the polypeptide may e.g. be recovered from the animal material, e.g. the milk, in which it is produced by extraction, centrifugation, affinity chromatography, ion exchange chromatography, gel filtration, or other conventionally used polypeptide isolation and purification techniques.
  • the recovery and isolation of the polypeptide will also of course depend on the kind of microorganism employed.
  • the recovering of the polypeptide from the microorganism comprises treatment of the microorganism so as to release the polypeptide, e.g. by rupturing the microorganism, i.e. partly or totally, and subsequently recovering the polypeptide by well-known methods such as precipitation, gel filtration, ion exchange chromatography, or HPLC reverse phase chromatography or immuno affinity chromatography or the like.
  • the polypeptide of the invention may be isolated from a biological material containing the polypeptide, e.g. a suspension of cells producing the polypeptide, by use of a method comprising adsorbing the biological material to a matrix comprising an immobilized monoclonal or polyclonal antibody as described herein, eluting the polypeptide from the matrix, and recovering the polypeptide from the eluate.
  • a biological material containing the polypeptide e.g. a suspension of cells producing the polypeptide
  • a method comprising adsorbing the biological material to a matrix comprising an immobilized monoclonal or polyclonal antibody as described herein, eluting the polypeptide from the matrix, and recovering the polypeptide from the eluate.
  • procedures for isolating the polypeptide are:
  • a procedure employing antibodies reactive with u-PAR compounds or with u-PAR-reactive compounds which is suited for the obtainment of a u-PAR containing fraction with high yield and purity.
  • the procedure may be performed by immobilizing the specific antibodies, preferably monoclonal antibodies, to a matrix, contacting said matrix with the preparation containing the released u-PAR compounds, washing, and finally treating the antigenantibody complex fixed to the matrix so as to release the u-PAR compounds in a purified form.
  • a preferred way is to isolate the u-PAR compounds by means of column affinity chromatography involving antibodies fixed to the column matrix.
  • Preparative electrophoresis procedures for instance the following procedure: A supernatant from a centrifuged enzyme treated cell or cell line preparation is subjected to a gel electrophoresis, such as a sodium dodecyl sulphate-polyacrylamidgel electrophoresis (SDS-PAGE) (cf. Laemmli, U.K. Nature, 227:680-685; 1970), or an agarose gel electrophoresis. Subsequently, labelled antibodies, such as monoclonal antibodies, reactive with u-PAR, are used to identify bands primarily constituted by the isolated u-PAR compounds. For instance, the antibodies may be used in any conventional immunoblotting technique.
  • a gel electrophoresis such as a sodium dodecyl sulphate-polyacrylamidgel electrophoresis (SDS-PAGE) (cf. Laemmli, U.K. Nature, 227:680-685; 1970), or an agarose
  • the markers may be isotopes or fluorescein labels detectable by means of relevant sensitive films.
  • the u-PAR containing bands of the gel may be subjected to a treatment resulting in the release of the u-PAR compounds from the gels, such as procedures involving slicing up the gel and subsequent elution of u-PAR compounds.
  • the amino acid sequence of the u-PAR proteins obtained may be determined.
  • the DNA fragment encoding the polypeptide of the invention may be subjected to modification, before or after the DNA fragment has been inserted in the vector.
  • the polypeptide produced may also be subjected to modification.
  • the modification may comprise substitution, addition, insertion, deletion or rearrangement of one or more nucleotides and amino acids in the DNA fragment and the polypeptide, respectively, or a combination of these modifications.
  • substitution is intended to mean the replacement of any one or more amino acids or nucleotides in the full amino acid or nucleotide sequence with one or more others
  • “addition” is understood to mean the addition of one or more amino acids or nucleotides at either end of the full amino acid or nucleotide sequence
  • “insertion” is intended to mean the introduction of one or more amino acids or nucleotides within the full amino acid or nucleotide sequence
  • detion is intended to indicate that one or more amino acids or nucleotides have been deleted from the full amino acid or nucleotide sequence whether at either end of the sequence or at any suitable point within it.
  • “Rearrangement” is intended to indicate that one or more amino acids or nucleotides or the sequence has been exchanged with each other.
  • the DNA fragment may, however, also be modified by subjecting the organism carrying the DNA fragment to mutagenization, preferably site directed mutagenization so as to mutagenize said fragment.
  • the mutagenization may be performed by using conventional mutagenization means such as ultraviolet radiation, ionizing radiation or a chemical mutagen such as mitomycin C, 5-bromouracil, methylmethane sulphonate, nitrogen mustard or a nitrofuran or mutagens known in the art, e.g. mutagens of the type disclosed in Miller, J. H., Molecular genetics, Unit III, Cold Spring Harbor Laboratory 1972.
  • nucleotide substitutions which do not give rise to another amino acid sequence of the protein, but which, e.g., correspond to the codon usage of the specific organism in which the sequence is inserted; nucleotide substitutions which give rise to a different amino acid sequence and therefore, possibly, a different protein structure without, however, impairing the critical properties of the polypeptide encoded by the DNA sequence; a subsequence of the DNA sequence shown above encoding a polypeptide which has retained the receptor properties of the native u-PAR; or a DNA sequence hybridizing to at least part of a DNA prepared on the basis of the DNA sequence shown above, provided that it encodes a polypeptide which has the biological property of u-PAR.
  • polypeptide produced as described above may be subjected to posttranslational modifications such as for instance thermal treatment, treatment with a chemical such as formaldehyde, glutar aldehyde or a suitable proteolytic enzyme, e.g. a peptidase or proteinase, such as trypsin, phospholipases, glycopeptidases.
  • a chemical such as formaldehyde, glutar aldehyde or a suitable proteolytic enzyme, e.g. a peptidase or proteinase, such as trypsin, phospholipases, glycopeptidases.
  • truncated polypeptide refers to a polypeptide deleted for one or more amino acid residues eventually resulting in changing of the properties of the polypeptide, e.g. solubility.
  • truncated polypeptide refers to a mixture of polypeptides all derived from one polypeptide or expressed from the gene encoding said polypeptide. Such truncated polypeptides might arise for instance in vector/host cell systems in which part of the cDNA has been deleted by restriction enzyme digestion or other suitable methods, resulting in the expression of a protein not normally produced in that system.
  • the solid support may also serve as the carrier for the polypeptide of the invention in a vaccine preparation as described below.
  • the preparation of synthetic peptides may be carried out essentially as described in Shinnick, Ann. Rev. Microbiol. 37, 1983, pp. 425-446.
  • Another aspect of the invention is a monoclonal or polyclonal antibody reactive with u-PAR compounds, and a method for the preparation thereof.
  • antibody refers to a substance which is produced by a vertebrate or more precisely a cell of vertebrate origin belonging to the immune system as a response to exposure to the polypeptides of the invention.
  • the variant domain of an antibody is composed of variable and constant sequences.
  • the variant part of the domain is called the idiotype of the antibody. This part of the antibody is responsible for the interaction with the antigen, the antigen binding.
  • the idiotypic structure is antigenic and can thus give rise to specific antibodies directed against the idiotypic structure. This has been done in mice.
  • the antibodies raised against the idiotype, the anti-idiotypic antibodies may mimic the structure of the original antigen and therefore may function as the original antigen to raise antibodies reactive with the original antigen.
  • This approach may be advantageous as it circumvents the problem associated with the characterizationand synthesis of the important immunogenic parts of the protein in question. This is most important in the case of conformational epitopes, which might otherwise be difficult to identify. It has been shown for a number of organisms that protective immunity can be induced in this way (e.g. Trypanosoma druzei, Trypanosoma brucei , Hepatitis B virus, and Plasmodium knowlesii ).
  • the antibodies of the present invention may be produced by a method which comprises administering in an immunogenic form at least a natural or synthetic part of the polypeptide of the invention to obtain cells producing antibodies reactive with said polypeptide and isolating the antibody containing material from the organism or the cells.
  • the methods of producing antibodies of the invention will be explained further below.
  • the antibody is preferably a monospecific antibody.
  • the monospecific antibody may be prepared by injecting a suitable animal with a substantially pure preparation of the polypeptide of the invention followed by one or more booster injections at suitable intervals (e.g. one or two weeks to a month) up to four or five months before the first bleeding. The established immunization schedule is continued, and the animals are bled about one week after each booster immunization, and antibody is isolated from the serum in a suitable manner (cf. e.g. Harboe and Ingild, Scand. J. Immun. 2 (Suppl. 1), 1973, pp. 161-164.)
  • the antibody may be a polyclonal antibody.
  • Polyclonal antibodies may be obtained, e.g. as described in Harboe and Ingild, see above. More specifically, when polyclonal antibodies are to be obtained, the u-PAR compound preparation is, preferably after addition of a suitable adjuvant, such as Freund's incomplete or complete adjuvant, injected into an animal. When the immunogens are human u-PAR compounds, the animals may be rabbits.
  • the animals are bled regularly, for instance at weekly intervals, and the blood obtained is separated into an antibody containing serum fraction, and optionally said fraction is subjected to further conventional procedures for antibody purification, and/or procedures involving use of purified u-PAR compounds.
  • monoclonal antibodies are obtained.
  • the monoclonal antibody may be raised against or directed substantially against an essential component of u-PAR compounds, i.e. an epitope.
  • the monoclonal antibody may be produced by conventional techniques (e.g. as described by Köhler and Milstein, Nature 256, 1975, p. 495) e.g. by use of a hybridoma cell line, or by clones or subclones thereof or by cells carrying genetic information from the hybridoma cell line coding for said monoclonal antibody.
  • the monoclonal antibody may be produced by fusing cells producing the monoclonal antibody with cells of a suitable cell line, and selecting and cloning the resulting hybridoma cells producing said monoclonal antibody.
  • the monoclonal antibody may be produced by immortalizing an unfused cell line producing said monoclonal antibody, subsequently growing the cells in a suitable medium to produce said antibody, and harvesting the monoclonal antibody from the growth medium.
  • the immunized animal used for the preparation of antibodies of the invention is preferably selected from the group consisting of rabbit, monkey, sheep, goat, mouse, rat, pig, horse and guinea pigs.
  • the cells producing the antibodies of the invention may be spleen cells or lymph cells, e.g. peripheric lymphocytes.
  • hybridoma cells When hybridoma cells are used in the production of antibodies of the invention, these may be grown in vitro or in a body cavity of an animal.
  • the antibody-producing cell is injected into an animal such as a mouse resulting in the formation of an ascites tumor which releases high concentrations of the antibody in the ascites of the animal.
  • the animals will also produce normal antibodies, these will only amount to a minor percentage of the monoclonal antibodies which may be purified from ascites by standard purification procedures such as centrifugation, filtration, precipitation, chromatography or a combination thereof.
  • the invention relates to a diagnostic agent capable of detecting and/or quantitating u-PAR or a derivative thereof in a sample.
  • such diagnostic agent may be valuable in diagnosis of cancer and other disorders involving tissue invasion and tissue remodelling, considering the involvement of u-PAR in these processes.
  • serum from breast cancer patients has an increased concentration of u-PA compared with normal individuals (Gr ⁇ ndahl-Hansen et al., 1988) and that the u-PA content in breast cancer tissue has been shown to be a valuable prognostic marker in this disease such as has been published in the priority year of the present application (Janicke et. al., 1989, 1990).
  • u-PAR u-PAE content in cancer tissue is an even better diagnostic and prognostic marker.
  • a new aspect of the potential diagnostic and prognostic use of u-PAR determinations is the release of u-PAR from cultured cells (described in Example 4) that occurs even in the absence of exogeneously added phospholipase. This finding raises the possibility that u-PAR is also released into body fluids under some physiological and pathophysiological conditions and particularly in cancer. Determination of concentrations of u-PAR or degradation products thereof in body fluids, such as serum, urine, and ascites fluid may therefore prove to be diagnostically and/or prognostically valuable.
  • the diagnostic agent may, e.g, be an antibody as defined above.
  • the diagnostic agent may be in the form of a test kit comprising in a container a polypeptide comprising a characteristic amino acid sequence of u-PAR, e.g. a sequence including or included in the sequence (1).
  • the diagnostic agent may be used in the diagnosis of diseases related to abnormal numbers of u-PARs residing on the cell.
  • the diagnostic agent may be one which is suited for use in an agglutination assay in which the solid particles to which the antibody is coupled agglutinate in the presence of a polypeptide of the invention in the sample subjected to testing. In this type of testing, no labelling of the antibody is necessary. For most uses it is, however, preferred that the antibody is provided with a label for the detection of bound antibody or, alternatively (such as in a double antibody assay), a combination of labelled and unlabelled antibody may be employed.
  • the substance used as label may be selected from any substance which is in itself detectable or which may be reacted with another substance to produce a detectable product. Thus, the label may be selected from radioactive isotopes, enzymes, chromophores, fluorescent or chemiluminescent substances, and complexing agents.
  • enzymes useful as labels are ⁇ -galactosidase, urease, glucose oxidase, carbonic anhydrase, peroxidases (e.g. horseradish peroxidase), phosphatases (e.g. alkaline or acid phosphatase), glucose-6-phosphate dehydrogenase and ribonuclease.
  • peroxidases e.g. horseradish peroxidase
  • phosphatases e.g. alkaline or acid phosphatase
  • glucose-6-phosphate dehydrogenase ribonuclease.
  • Enzymes are not in themselves detectable, but must be combined with a substrate to catalyze a reaction the end product of which is detectable.
  • a substrate may be added to the reaction mixture resulting in a coloured, fluorescent or chemiluminescent product or in a colour change or in a change in the intensity of the colour, fluorescence or chemiluminescence.
  • substrates which are useful in the present method as substrates for the enzymes mentioned above are H 2 O 2 , p-nitrophenylphosphate, lactose, urea, ⁇ -D-glucose, CO 2 , RNA, starch, or malate.
  • the substrate may be combined with, e.g. a chromophore which is either a donor or acceptor.
  • Chromophores may be o-phenylenediamine or similar compounds. These substances may be detected by means of a spectrophotometer. Radioactive isotopes may be any detectable and in a laboratory acceptable isotope, e.g. 125 I, 131 I, 3 H, 35 P, 35 S or 14 C. The radioactivity may be measured in a ⁇ -counter or a scintillation counter or by radio-autography followed by densitometry.
  • Complexing agents may be Protein A, Protein G (which form a complex with immunoglobulins), biotin (which forms a complex with avidin and streptavidin), and lectin (which forms a complex with carbohydrate determinants, e.g. receptors).
  • the complex is not in itself directly detectable, necessitating labelling of the substance with which the complexing agent forms a complex.
  • the marking may be performed with any of the labelling substances described above.
  • an antibody or a polypeptide of the invention may be coupled to a bridging compound coupled to a solid support.
  • the bridging compound which is designed to link the solid support and the antibody may be hydrazide, Protein A, glutaraldehyde, carbodiimide, or lysine.
  • the solid support employed is e.g. a polymer or it may be a matrix coated with a polymer.
  • the matrix may be of any suitable solid material, e.g. glass, paper or plastic.
  • the polymer may be a plastic, cellulose such as specially treated paper. nitrocellulose paper or cyanogenbromide-activated paper.
  • suitable plastics are latex, a polystyrene, polyvinylchloride, polyurethane, polyacrylamide, polyvinylacetate and any suitable copolymer thereof.
  • silicone polymers include siloxane.
  • the solid support may be in the form of a tray, a plate such as a mitrotiter plate, e.g. a thin layer or, preferably, strip, film, threads, solid particles such as beads, including Protein A-coated bacteria, or paper.
  • a mitrotiter plate e.g. a thin layer or, preferably, strip, film, threads, solid particles such as beads, including Protein A-coated bacteria, or paper.
  • the polypeptide and antibody of the invention may be used in an assay for the identification and/or quantification of at least a form and/or a part of said polypeptide present in a sample.
  • the identification and/or quantification performed by the use according to the present invention may be any identification and/or quantification involving u-PAR compounds or a form of u-PAR compounds. Thus, both a qualitative and a quantitative determination of u-PAR compounds may be obtained according to the use of the present invention.
  • the identification and/or quantification may be performed for both a scientific, a clinical and an industrial purpose. As will be further described below, it is especially important in clinical routine to identify or quantify u-PAR compounds.
  • the sample may be a specimen obtained from a living organism such as a human or an animal.
  • the specimen may be blood, e.g. an erythrocyte enriched fraction, or a tissue sample e.g. comprising liver cells.
  • the specimen is urine.
  • the antibody used in the method of the invention is a monoclonal antibody as this generally provides a higher precision and accuracy of the assay, at the same time possibly requiring less time to perform. Furthermore, a mixture of two or more different monoclonal antibodies may be employed as this may increase the detection limit and sensitivity of the test.
  • the monoclonal antibody may be obtained by the method described below. Antibodies possessing high avidity may be selected for catching techniques.
  • the antibody used in the present method is preferably in substantially pure form (purified according to suitable techniques or by the methods of the invention, see below) in order to improve the precision and/or accuracy of the assays of the invention.
  • the determination of antibodies reactive with the polypeptide of the invention and being present in a sample may be carried out by use of a method comprising contacting the sample with the polypeptide of the invention and detecting the presence of bound antibody resulting from said contacting and correlating the result with a reference value.
  • the polypeptide of the invention When the polypeptide of the invention is to be employed in an assay for determining the presence of u-PAR compounds in a sample, it may be in the form of a diagnostic reagent or a diagnostic agent. As will be apparent to a person skilled in the art several techniques may be applied in connection with such diagnostic reagents.
  • any part of said polypeptide is coupled to a solid support
  • an antibody against the component may then be added to the solid support.
  • the antibody is coupled to a solid support.
  • any u-PAR compounds present in the sample is coupled to a solid support. It may then be incubated with the polypeptide component by addition of the component to the solid support followed by adding an antibody labelled with a detectable marker.
  • PCR polymerase chain reaction
  • the oligonucleotide primers are constructed so as to enable hybridization of one primer to the plus strand 5′ of the target DNA, and of another primer to the minus strand 5′ of the Target DNA.
  • the preferred distance between the two primers is 500-2000 base pairs for diagnostic purposes, whereas longer distances could be accepted for preparative purposes.
  • the primers are hybridized with the opposite DNA strands to be amplified and are extended by using DNA polymerase, e.g. the Klenow fragment of E.
  • DNA polymerase I or another useful DNA polymerase such as the Taq DNA polymerase, so as to synthesize a DNA sequence which is complementary to the DNA sequence to which the primers are annealed.
  • the DNA synthesized is denatured, e.g. by heating, from the “parent DNA strings”, and the parent strings as well as the newly synthesized DNA strings are subjected to a new PCR amplification cycle. In this manner, it is possible to obtain a substantial amplification of specific DNA sequences which are present in a sample.
  • the PCR amplification method it may be possible to amplify and thus detect the presence of originally very small and undetectable amounts of DNA sequences present in a sample, and thereby e.g. identifying a cancer cell.
  • FIGS. 1A-C SDS-PAGE of affinity-purified u-PAR and chemical cross-linking to specific ligands.
  • FIG. 1A The Triton X-114 fraction containing membrane proteins from PMA-treated U937a cells was subjected to affinity chromatography using immobilized DFP-treated u-PA. The neutralized column eluate was dialyzed aginst 0.1% acetic acid and concentrated by lyophilization. A portion, representing 2 ⁇ 10 8 cells before purification, was run on 6-16% gradient SDS-PAGE under reducing conditions (lane 1). The gel was silver-stained. The molecular weights of marker proteins (lane 2) are indicated.
  • FIG. 1B Affinity column eluate was diluted to yield an approximate concentration of u-PAR of 1 nM during the assay.
  • the samples were preincubated alone (lane 2) or in the presence of the following unlabelled reagents at a concentration of 100 nM: bovine serum albumin (lane 3), t-PA (lane 4), plasminogen (lane 5), murine epidermal growth factor (lane 6), ATF (lane 7), active 54 kD u-PA (lane 8), DFP-inactivated 54 kD u-PA (lane 9).
  • bovine serum albumin lane 3
  • t-PA lane 4
  • plasminogen lane 5
  • murine epidermal growth factor lane 6
  • ATF lane 7
  • active 54 kD u-PA lane 8
  • DFP-inactivated 54 kD u-PA lane 9
  • Lane 1 shows the cross-linked control with 125 I-ATF and no addition of u-PAR or competitors. Electrophoretic mobilities of molecular weight standard proteins are inducated (kD).
  • FIG. 1C Neutralized affinity column eluate was concentrated as in (A) to yield a u-PAR concentration of approximately 15 ⁇ g/ml, and subjected to cross-linking with DSS in the presence of 50 ⁇ g/ml DFP-treated u-PA (lane 3) or alone (lane 4).
  • the samples were run on Phast-SDS-PAGE under non-reducing conditions. Each lane contained 10 ng of u-PAR and/or 33 ng of DFP-u-PA.
  • the gel was silver-stained. Note that the chemical cross-linking led to a minor increase in the migration rate of DFP-treated u-PA alone, probably due to internal cross-binding, but not of u-PAR alone. Electrophoretic mobilities of molecular weight standard proteins (lane 6) are indicated (kD).
  • FIG. 2 Enzymatic deglycosylation of purified u-PAR.
  • Affinity purified 125 I-labelled u-PAR was pretreated for deglycosylation by denaturation under mildly reducing conditions (see “Experimental Procedures”) and treated with peptide:N-glycosidase F (lane 2) or analyzed directly (lane 1).
  • Analysis was performed by SDS-PAGE under reducing conditions on a 6-16% gradient gel, followed by autoradiography on a Kodak XAR film. Electrophoretic mobilities of standard proteins are indicated (kD).
  • FIG. 3 Deglycosylation of cross-linked 125 I-ATF: u-PAR complexes from PMA-treated and nontreated U937a cells.
  • PMA-treated (lanes 1 and 3) and nontreated (lanes 2 and 4) cells were acid-treated and lysed with 0.5% CHAPS.
  • the lysates were incubated with 125 I-ATF, cross-linked with disuccinimidyl suberate, denatured under mildly reducing conditions, and then further incubated in the presence (lanes 3 and 4) or absence (lanes 1 and 2) of peptide:N-glycosidase F, and analyzed by SDS-polyacrylamide (6-16%) gel electrophoresis under reducing conditions, followed by autoradiography. Electrophoretic mobilities of standard proteins are indicated (kD).
  • FIG. 4 shows direct electrophoretic analysis of chymotryptic fragments of u-PAR.
  • Samples of purified u-PAR were degraded at 37° C., using the following, final concentrations of chymotrypsin: 8 ng/ml (lane 1), 40 ng/ml (lane 2) or 200 ng/ml (lane 3), or incubated under the same conditions without the addition of enzyme (lanes 4 and 5). After 7 h of incubation (lanes 1-4) or immediately, without incubation (lane 5), phenylmethylsulfonylfluoride (1 mM final concentration) was added. The samples were analyzed by Tricine-SDS-PAGE under reducing conditions, followed by silver staining. The electrophoretic mobilities of molecular weight marker proteins are indicated (kD).
  • FIG. 5 shows chymotryptic fragments of u-PAR, analyzed by chemical cross-linking to 125 I-ATF. Preparation of samples and numbering of lanes are the same as in FIG. 4 . The samples were 50-fold diluted and analyzed by chemical cross-linking to 125 I-ATF, followed by SDS-PAGE on a 6-16% gradient gel under reducing conditions, and autoradiography. The electrophoretic mobilities of molecular weight marker proteins are indicated (kD).
  • FIG. 6 shows deglycosylation of chymotryptic fragments, cross-linked to ATF.
  • Samples of purified u-PAR were subjected to degradation with 8 ng/ml chymotrypsin (lanes 1 and 4) or 40 ng/ml chymotrypsin (lanes 2 and 5) as described in the legend to FIG. 4, including termination with phenylmethylsulfonylfluoride.
  • the samples in lanes 3 and 6 received no enzyme and were not incubated but received the same amount of phenylmethylsulfonylfluoride.
  • the samples were 50-fold diluted, and subjected to chemical cross-linking to 125 I-ATF.
  • the cross-linked samples were subjected to enzymatic deglycosylation with N-Glycanase (lanes 4-6) or treated in parallel without the addition of N-Glycanase (lanes 1-3).
  • Analysis was performed by SDS-PAGE on a 6-16% gradient gel under reducing conditions, followed by autoradiography. The electrophoretic mobilities of molecular weight marker proteins are indicated (kD).
  • FIGS. 7A-C show the initial amino-terminal amino acid sequence (SEQ ID NO: 24) information and the oligonucleotide synthesized and used for library screening (SEQ ID NOS: 25 and 26); I stands for inosine.
  • FIG. 7B shows the restriction map of p-uPAR-1 clone and the strategy employed for the complete double stranded sequence.
  • FIG. 7C shows the hydrophobicity plot. The abscissa shows the amino acid residue position, the ordinate the degree of hydrophobicity calculated using the algorithm of Hopp and Wood (1981) and Kyte and Doolittle (1982).
  • FIG. 8 shows a map of the plasmid p-uPAR-1.
  • the plasmid was isolated from the Okayama-Berg cDNA library by hybridization to the 24-mer oligonucleotide constructed from the N-terminal peptide.
  • FIGS. 9A-9F Caseinolytic plaque assay of uPA binding to LB6 cells transfected with p-uPAR-1 DNA.
  • FIGS. 9 A and C-F refer to clone LB6/p-uPAR-1 while plate B refers to clone LB6/RSVCAT. In plate A no uPA was added. Otherwise (FIGS. 9B-F) cells were subjected to a binding step with 0.2 nM human uPA for 1 hour at 37° C. The following competitors, present during the binding step, were used: none (FIGS. 9B, 9 C); 100 nM ATF (FIG. 9 D); 200 ⁇ M synthetic peptide human uPA[12-32(ala19)] (FIG. 9 E); 100 ⁇ M synthetic peptide mouse uPA[13-33(ala20)] (FIG. 9 F).
  • FIG. 10 A Binding of human 125 I-ATF to mouse LB6 cells transfected with RSVCAT (closed circles) and p-uPAR-1 DNA (closed circles). Specific binding was calculated by subtracting the counts not competed by 100 nM unlabelled ATF (about 1000 cpm in this experiment).
  • FIG. 10 B Reducing SDS-polyacrylamide (12.5%) gel electrophoretic analysis of the 125 I-ATF cross-linked to LB6/p-uPAR-1 cells.
  • Lane 1 has the molecular weight markers (see Methods); lane 2 represents the migration of the labelled ATF (3,000 cpm).
  • Lanes 3 and 4 show the migration of duplicate LB6/p-uPAR-1 extracts cross-linked with ligand.
  • Lanes 5 and 6 show the competition of the cross-linking of LB6/p-uPAR-1 cells to the ligand by unlabelled ATF (100 nM final concentration). The last lane to the right shows the cross-linking obtained (in a separate experiment) with the same ligand and the human GM637 cells which served as a source of RNA for the cDNA library used to isolate p-uPAR-1.
  • FIG. 11 shows SDS-PAGE (12.5%) electrophoretic analysis of the p-u-PAR-PFLM-1 mutant transfected into LB6 cells.
  • Cells were incubated with iodinated ATF, washed, extracted with Triton X-114, and an amount of extract corresponding to 300,000 cells cross-linked with DSS as described before and run on the gel (part C of the Figure).
  • conditioned medium was centrifuged at 100,000 ⁇ g, and the supernatant (a volume corresponding to 15,000 cells) was incubated with iodinated ATF, cross-linked with DSS, and analyzed by SDS-PAGE (part B of the Figure).
  • Lanes a and b are duplicates from cells grown at different densities.
  • FIGS. 12A-12B show an elution profile from cation-exchange chromatography of amino acids released from u-PAR after acid hydrolysis.
  • the protein was initially purified from PMA-stimulated U937 cells (6 ⁇ 10 9 cells) by Triton X-114 detergent-phase separation and affinity chromatography (DFP-u-PA Sepharose). To improve purity and eliminate interference on amino acid analysis from low molecular weight compounds, this receptor preparation was dialysed thoroughly against 0.1% acetic acid, lyophilized and then subjected to Tricine-SDS-PAGE followed by electrotransfer onto a 0.45 ⁇ m PVDF-membrane (8 cm ⁇ 8 cm).
  • the insert shows the immobilized u-PAR after staining with Coomassie Brilliant Blue R-250.
  • a slight decrease in mobility of u-PAR was observed in this experiment, due to a large excess of the zwitterionic detergent CHAPS in the lyophilized preparation.
  • the stained area of the PVDF-membrane representing u-PAR was excised and hydrolysed in vacuo for 20 hours at 110° in the presence of 3,3′-dithiodipropionic acid (DTDPA). Cys-X is the product formed between cysteine and DTDPA during hydrolysis, GlcN is glucosamine and EtN is ethanolamine.
  • FIG. 13 shows the release of 125 I-labelled DFP-treated u-PA from adherent, PMA-stimulated U937 cells by phosphatidylinositol-specific phospholipase C from Bacillus cereus (PI-PLC).
  • PI-PLC phosphatidylinositol-specific phospholipase C from Bacillus cereus
  • FIGS. 14A-C show complex formation and molecular analysis by SDS-PAGE of 125 I-labelled DFP-treated u-PA and u-PAR released to the medium by PI-PLC.
  • Aliquots of supernatants from the experiment described in FIG. 13 were analysed by SDS-PAGE (10% T, 2.5% C) under non-reducing conditions either directly (FIG. 14A) or subsequent to cross-linking with 1 mM disuccinimidyl suberate (DSS), performed immediately after sampling (FIG. 14 B).
  • SDS-PAGE 10% T, 2.5% C
  • DSS disuccinimidyl suberate
  • FIG. 14 C 2 dishes of PMA-stimulated U937 cells were cultured and acid treated as described in the legend to FIG. 13 .
  • one dish was incubated at 37° C. in 5 ml of serum-free medium (RPMI 1640 including 25 mM HEPES, pH 7.4) with 0.6 ⁇ g PI-PLC, whereas the other was incubated in 5 ml of medium only. Aliquots were withdrawn at 0 min, 30 min and 60 min after the addition of lipase and centrifuged immediately (20,000 ⁇ g for 5 min). Supernatants were preincubated for 1 hour at 4° C. with 125 I-labelled DFP-inactivated u-PA (1 nM) and then cross-linked with 1 mM DSS. The rightmost lane (DFP-u-PA) represents the 125 I-labelled ligand cross-linked in the absence of u-PAR. Samples were analysed by SDS-PAGE as above.
  • FIGS. 15A-15C show the change in hydrophobic properties of purified u-PAR upon treatment with PI-PLC.
  • u-PAR purified from PMA-stimulated U937 cells, were either untreated (NONE) FIG. 15A or incubated for 30 min at 37° C. in 50 mM triethylamine/HCl (pH 7.5), 5 mM EDTA and 0.1% Triton X-100 without any phospholipases (MOCK) FIG. 15B or in the presence of 20 ⁇ l/ml PI-PLC (PI-PLC) FIG. 15 B.
  • FIG. 16 shows a comparison of COOH-terminal amino acid sequences from proteins, in which the processing sites during GPI-membrane anchoring are known, to that predicted for u-PAR (SEQ ID NO: 21) (based on amino acid analysis, Table 5). The amino acids involved in attachment to the glycolipid are highlighted.
  • VSG SEQ ID NO: 17
  • PARP SEQ ID NO: 16
  • VSG SEQ ID NO: 17
  • PARP SEQ ID NO: 16
  • CEA (SEQ ID NO: 19) is carcinoembryonic antigen; PLAP (SEQ ID NO: 18) is placental alkaline phosphatase and Thy-1 (SEQ ID NO: 20) refers to the surface glycoprotein isolated from rat thymocytes.
  • FIGS. 17A-B show Northern blot (FIG.S 17 A and 17 B) analysis of u-PAR mRNA in U937 cells treated with PMA.
  • the cells were seeded at 0.5 ⁇ 10 6 /ml in RPMI 1640 medium containing 10% FCS, and incubated with 150 nM PMA for the indicated number of hours. After the incubation both the adherent and the non-adherent cells were collected, pooled, and total RNA was isolated as described in Materials and Methods.
  • For the Northern blot analyses 30 ⁇ g of total RNA were electrophoresed in 1.5% agarose gels under denaturing conditions and blotted onto a nitrocellulose filter.
  • the filters were hybridized to a random primed labelled u-PAR cDNA probe (FIG. 17 A), or a ⁇ -actin cDNA probe (FIG. 17 B). The positions of the ribosomal RNA in the Northern blot are indicated to the left.
  • FIG. 18 shows SDS-PAGE of detergent phase from Triton X-114 phase-separated extracts from U937 cells treated with PMA for different time periods, chemical cross linked to 125 I-ATF.
  • Non-treated cells and PMA (150 nM) treated cells were acid treated and lysed.
  • the detergent phases were incubated with 125 I-ATF, cross linked with DSS and run in a 6-16% SDS-PAGE gradient gel followed by autoradiography. Electrophoretic mobility of molecular weight standard proteins are indicated to the left. 1.
  • Non-treated cells 2. +PMA 3 hours, 3. +PMA 9 hours, 4. +PMA 24 hours, 5. +PMA 48 hours, 6. Blind, 7. 1% Triton X-114 total lysate (diluted 1/25) from HEp2 cells.
  • FIG. 19 shows Northern blot analysis of u-PAR mRNA in A549 and RD cells treated with PMA, dexamethasone, mEGF and pTGF- ⁇ 1.
  • the cells were seeded at 0.5 ⁇ 10 6 /ml in DMEM and grown to confluence, washed in PBS + and kept under serum-free conditions for 48 hours followed by no additions (1, 6), by addition of Dex (10 ⁇ 6 M) (2, 7) PMA (150 nM) (3, 8) mEGF (20 ng/ml) (4, 9) and TGF- ⁇ 1, (7.5 ng/ml) (5, 10) for 48 hours.
  • 30 ⁇ g of total RNA were electrophoresed in 1.5% agarose gels under denaturing conditions and blotted onto a nitrocellulose filter. The filters were hybridized to a random primed labeled u-PAR cDNA probe. The positions of the ribosomal RNA in the Northern are indicated to the left.
  • FIG. 20 shows SDS-PAGE of detergent phase from Triton X-114 phase-separated extracts from U937 cells treated with Dibuturyl cAMP for different time periods, chemical cross-linked to 125 I-ATF.
  • Non-treated cells and Dibuturyl cAMP (1 mM) treated cells were acid treated and lysed as described in Materials and Methods.
  • the detergent phases were incubated with 125 I-ATF, cross linked with DSS and run in a 6-16% SDS-PAGE gradient gel followed by autoradiography. Electrophoretic mobility of molecular weight standard proteins are indicated to the left. 1.
  • Non-treated cells 2. +Dibuturyl cAMP 12 hours, 3. +Dibuturyl cAMP 24 hours, 4. +Dibuturyl cAMP 48 hours, 5. +Dibuturyl cAMP 72 hours.
  • FIGS. 21A-E show hybridization to paraffin sections of human colon adenocarcinoma using antisense RNA generated from the cDNA subclone pHUR06.
  • Disrupted tumor glands at invasive foci show hybridization to cells at the leading edge of strands of tumor cells (arrows in FIG. 21B; FIG. 21B is a magnification of squared area in FIG. 21 A).
  • hybridization signal is located above cells at the abluminal surface of the malignant epithelium (FIG. 21C; arrows), or above cells located in stromal tissue surrounding the gland (FIG. 21D; arrow).
  • FIG. 21 E In areas of neovascularization cells of seemingly mesenchymal origin show hybridization (FIG. 21 E). Magnifications: 216 ⁇ (FIG. 21 A), 540 ⁇ (FIGS. 21 B-D), 870 ⁇ (FIGS. 21 E).
  • FIG. 22 Dependence of plasmin formation in serum medium on the concentration of added native human plasminogen.
  • Confluent layers of HT-1080 cells were incubated for 3 hours in MEM medium (0.5 ml) containing 10% heat-inactivated and plasminogen-depleted fetal calf serum, with the addition of native human plasminogen to the concentrations shown.
  • the conditioned media were harvested and the cells rinsed three times with PBS.
  • the cells were treated with 1 mM tranexamic acid in PBS to obtain the bound fraction of plasmin. Plasmin was assayed in the cell-bound fraction ( ⁇ -- ⁇ ) and the medium ( ⁇ -- ⁇ ) as thioesterase activity.
  • FIG. 23 Plasmin uptake onto HT-1080 cells from serum-free and serum-containing media.
  • Human plasmin (0-5 ⁇ g/ml) was added to HT-1080 cell layers growing in either serum-free MEM medium (SFM, 0.5 ml) ( ⁇ -- ⁇ or ⁇ -- ⁇ ) or MEM medium with 10% heat-inactivated fetal calf serum (SM) ( ⁇ -- ⁇ or ⁇ -- ⁇ ).
  • SFM serum-free MEM medium
  • SM heat-inactivated fetal calf serum
  • the plasmin in the bound fraction (A) and the conditioned media (B) were assayed as thioesterase activity. Note the different scales used in FIG. 23B for activity in SFM & SM.
  • FIG. 24 Plasmin release from HT-1080 cells into serum-free and serum-containing media. Confluent layers of HT-1080 cells were first loaded with plasmin by incubation for 1 hour at 37° C. in serum-free MEM medium (0.5 ml) containing human plasmin (0-5 ⁇ g/ml). After rinsing the cell layers three times, they were incubated for 2 hours at 37° C. with either serum-free medium ( ⁇ -- ⁇ ), medium containing 10% heat-inactivated and plasminogen-depleted fetal calf serum ( ⁇ -- ⁇ ), or the latter with tranexamic acid (100 ⁇ M) ( ⁇ -- ⁇ ). Plasmin was then assayed in the cell-bound fraction (A) and the media (B). At the time of transfer to new media, there was approximately 28 ng of plasmin bound to the cells from the pretreatment with 2.5 ⁇ g of plasmin/well.
  • FIG. 25 Effect of pretreatment of HT-1080 cells with DFP-u-PA on bound u-PA activity and ability to produce bound plasmin in serum medium.
  • Confluent cell layers of HT-1080 cells were preincubated for 18 hours at 37° C. with the concentrations shown of DFP-u-PA in serum-containing medium (0.5 ml). After rinsing three times, the cells were incubated for 1 hour at 37° C. with MEM medium containing 10% heat-inactivated and plasminogen-depleted fetal calf serum, with addition of native human plasminogen (40 ⁇ g/ml).
  • FIG. 26 Activation of cell-bound u-PA proenzyme in serum medium after addition of plasminogen.
  • Confluent layers of HT-1080 cells were prelabelled for 5 hours at 37° C. with 35 S-methionine.
  • native human plasminogen 50 ⁇ g/ml
  • Aprotinin 200 KIU/ml
  • Acid eluates were neutralized and immunoprecipitated with goat antibodies to u-PA, before SDS-PAGE under reducing conditions.
  • the fluorogram shows: in lane 1, control immunoprecipitate of culture without plasminogen with goat antibodies to human t-PA; lane 2, culture without plasminogen immunoprecipitated with goat anti-u-PA antibodies; lane 3, culture with plasminogen immunoprecipitated with u-PA antibodies.
  • FIG. 27 Activation of cell-bound u-PA proenzyme in serum medium after the addition of plasminogen.
  • Confluent layers of HT-1080 cells were incubated with MEM medium containing 10% heat-inactivated and plasminogen-depleted fetal calf serum and native human plasminogen (40 ⁇ g/ml).
  • aprotinin 200 KIU/ml was added and the rinsed cells were treated with acid-glycine to recover the bound fraction of u-PA.
  • the u-PA in the neutralized eluate was assayed by an immunocapture method, using an NPGB inactivation step to determined the pro-u-PA index (see Methods).
  • FIG. 27 Activation of cell-bound u-PA proenzyme in serum medium after the addition of plasminogen.
  • Confluent layers of HT-1080 cells were incubated with MEM medium containing 10% heat-inactivated and plasminogen-depleted fetal calf
  • FIG. 27A shows the pro-u-PA index for cultures without ( ⁇ -- ⁇ ) and with ( ⁇ -- ⁇ ) plasminogen.
  • the zero-time sample with plasminogen shows that some change already occurred during work-up of the cells.
  • FIG. 27B shows the eluted u-PA activity from cultures without plasminogen ( ⁇ -- ⁇ ), with plasminogen ( ⁇ -- ⁇ ), and with plasminogen and a neutralizing monoclonal antibody to human PAI-1 (10 ⁇ g/ml) ( ⁇ -- ⁇ ).
  • FIG. 28 Model for cell surface plasminogen activation.
  • u-PA-R u-PA receptors
  • plg-R plasminogen receptors
  • FIG. 28 Model for cell surface plasminogen activation.
  • u-PA-R u-PA receptors
  • plg-R plasminogen receptors
  • This step may be inhibited by PAI-1 and PAI-2, and by an anti-catalytic monoclonal antibody to u-PA (anti-u-PA-ab).
  • the bound plasmin thus formed is resistant to inhibition by the alpha-2-anti-plasmin present in the serum medium, but sensitive to inhibition by aprotinin and an anti-catalytic monoclonal antibody to plasmin (anti-pl-ab).
  • active plasmin As active plasmin becomes available, it catalyzes the activation of more bound pro-u-PA to active u-PA, thus amplifying the proteolytic system.
  • Activation of pro-u-PA is inhibited by tranexamic acid (which prevents plasminogen binding), aprotinin and an anti-catalytic monoclonal antibody to plasmin.
  • FIGS. 29A-29B Competition by unlabelled u-PA ( ⁇ -- ⁇ ) or u-PA/PAI-1 complex ( ⁇ -- ⁇ ) of the binding of 125 I-ATF to human U937 cells.
  • [Competitor] is the concentration of free or PAI-1 complexed u-PA; for PAI-1/u-PA complex formation, a 50 fold excess of PAI-1 was preincubated with u-PA for 1 hour at room temperature. Receptor binding was carried out at 4° C. for 90 minutes.
  • FIG. 29B shows the zymographic analysis of u-PA and PAI-1/u-PA solutions before (lanes 3 and 6) and after (lanes 1, 2, 4, 5) incubation with U937 cells.
  • FIG. 30 SDS-PAGE analysis of the binding of 125 I-u-PA/PAI-1 complexes to U937 cells.
  • the three leftmost lanes show the labelled u-PA preparation used for binding, before and after complexing with a 50 or 150 fold PAI-1 excess.
  • On top of each lane the identity of unlabelled competitor and the fold excess of PAI-1 over 125 I-u-PA is indicated.
  • the arrows to the right show the migration of high molecular weight (HMW), low molecular (LMW) u-PA and of the u-PA/PAI-1 complex.
  • the numbers to the left indicate the migration of molecular weight markers.
  • FIG. 31 SDS-PAGE analysis of binding of PAI-1 to receptor-bound u-PA in U937 cells.
  • the lane marked pro-u-PA shows the 125 I-pro-u-PA used for the binding.
  • Lane C shows the analysis of cell extracts incubated with non-activated, iodinated pro-u-PA only. Numbers on top indicate the nM concentration of PAI-1 in the binding mixtures.
  • the two sets of 4 lanes each marked “Binding's supern.” and “cell-bound” refer to the analysis of supernatants and cells.
  • the set of 4 lanes marked “solution” show the result obtained in complexing activated pro-u-PA with PAI-1 in the absence of cells.
  • FIGS. 32A-G Caseinolytic plaque assay of PAI-1 inhibition of receptor-bound u-PA.
  • FIGS. 32 A-D cells were incubated for 1 hour at 4° C. with: FIG. 32 A: 0.1% BSA.
  • FIG. 32 B 10 nM u-PA in 0.1% BSA.
  • FIG. 32 C 10 nM DFP-u-PA in 0.1% BSA.
  • FIG. 32 D 10 nM u-PA plus 750 nM PAI-1 in 0.1% BSA.
  • FIGS. 32 E-G cells were subjected to two serial incubations, first for 60 minutes at 4° C., then washed and reincubated at room temperature for further 20 minutes:
  • FIG. 32 E first with 10 nM DFP-u-PA in BSA and then with 10 nM u-PA in BSA.
  • FIG. 32 F first with 10 nM u-PA in BSA and then with 10 nM DFP-u-PA and 750 nM PAI-1.
  • FIG. 32 G first with 10 nM u-PA in BSA and then with 10 nM DFP-u-PA.
  • the cells were washed thoroughly, overlaid with agar containing casein and plasminogen (see Methods section), and incubated for 3 hours at 37° C.
  • FIGS. 33A-D show the fate of receptor-bound ligands after switch of the temperature from 4° C. to 37° C.
  • U937 cells were incubated for 90 minutes at 4° C. with the indicated ligands, and the cells were then washed and incubated at 37° C. in the absence of ligands for the indicated times. The data are expressed in percentage of the total counts recovered at time zero of the step 2 incubation.
  • Open circles receptor-bound ligand.
  • Full circles cell-associated, non-acid-extractable ligand.
  • Open triangles TCA-soluble radioactivity in the supernatant. Receptor-bound and cell-bound radioactivity were always totally precipitable with TCA.
  • FIG. 34 shows the fate of receptor-bound u-PA in the presence of 50 nM low molecular weight u-PA during the whole experiment (step 1, step 2, and washing buffers).
  • Open symbols represent total radioactivity, filled-in symbols represent TCA-precipitable counts.
  • Squares receptor-bound u-PA (i.e. acid-extracted).
  • Triangles radioactivity in the supernatants.
  • Circles non-acid-extractable cell-associated u-PA.
  • the ordinate (cpm, %) is expressed in percent of the counts recovered at the end of step 1 incubation.
  • FIGS. 35A-B show the effect of chloroquine treatment on u-PA:PAI-1 degradation by U937 cells.
  • FIG. 35 A Control (no addition).
  • FIG. 35 B 0.5 mM chloroquine during both step 1 and step 2 incubations. Open circles: receptor-bound ligand. Triangles: non-acid-extractable, cell-associated ligand. Filled-in circles: degraded ligand in the supernatant. The ordinate (cpm, %) is expressed in percent of the counts recovered at the end of step 1 incubation. The total counts should therefore add to 100%. The missing amount represents undegraded ligand present in the supernatant (see insert). In all cases, both receptor-bound and cell-associated, non-acid-extractable ligand were 100% precipitable by TCA.
  • FIG. 36 shows plasminogen activation by uPAR-bound u-PA on U937 cells.
  • Data are shown for U937 washed in buffer alone i.e. with endogenously-bound u-PA ( ⁇ ), cells pre-incubated with u-PA ( ⁇ ), cells acid-washed and then pre-incubated with u-PA ( ⁇ ), acid-washed cells ( ⁇ ) and cells pre-incubated with an anti-catalytic monoclonal antibody to u-PA (10 ⁇ g/ml for 60 minutes) ( ⁇ ).
  • the data shown are from single incubations, and are representative of data obtained from at least quadruplicate determinations.
  • FIG. 37 shows the inhibition of uPAR-bound u-PA by PAI-1.
  • PAI-1 concentrations were 0.18 nM ( ⁇ ), 0.46 nM ( ⁇ ), 1.84 nM ( ⁇ ), 4.60 nM ( ⁇ ) and 18.4 nM ( ⁇ ). Plasmin generation in the absence of PAI-1 is also shown ( ⁇ ).
  • the lines drawn represent the best fit of the experimental data at each inhibitor concentration to equation 1 (see Materials and Methods) by non-linear regression analysis, from which k app's were calculated.
  • the experimental data shown are from single incubations at each inhibitor concentration, which were representative of the data obtained from quadruplicate determinations.
  • FIG. 38 shows the inhibition of uPAR-bound u-PA by PAI-2.
  • PAI-2 concentrations were 1.13 nM ( ⁇ ), 5.67 nM ( ⁇ ), 11.3 nM ( ⁇ ), 28.3 nM ( ⁇ ) and 56.7 nM ( ⁇ ). Plasmin generation in the absence of PAI-2 is also shown ( ⁇ ). The lines were drawn as described in the legend to FIG. 2 . The experimental data shown are from single incubations at each inhibitor concentration, which were representative of the data obtained from triplicate determinations.
  • FIG. 39 shows a comparison of the inhibition of uPAR-bound and fluid-phase u-PA by PAI-1 and PAI-2.
  • k app 's determined from equation 1 were plotted against inhibitor concentration in a double-reciprocal manner. Association rate constants were calculated from the reciprocal of the slopes of these lines, for the inhibition of receptor-bound u-PA (open symbols) and u-PA in solution (closed symbols) by PAI-1 ( ⁇ , ⁇ ) and PAI-2 ( ⁇ , ⁇ ).
  • the data points shown are the means of at least triplicate determinations.
  • FIG. 40 shows the kinetics of plasminogen activation by u-PA bound to u-PAR on U937 cells.
  • U937 cells pre-incubated with u-PA, which was demonstrated to be specifically bound to u-PAR by competition with DFP-u-PA and anti-u-PAR antibodies, were incubated with Glu-plasminogen (0.09-2.26 ⁇ M). Rates of plasmin generation were plotted against plasminogen concentrations in a double-reciprocal manner.
  • K m was determined as 0.67 ⁇ M and V max as 0.043 nM min ⁇ 1 which, at an experimentally determined cell-bound u-PA concentration of 7.7 pM, is equivalent to a k cat of 5.6 min ⁇ 1 .
  • FIG. 41 shows the effect of u-PAR, released from PMA-stimulated U937 cells by PI-PLC, on u-PA activity.
  • PMA-stimulated U937 cells were treated with PI-PLC for 120 minutes, then pre-incubated with or without antibody to PAI-2. Residual plasmin generation by u-PA is shown after incubation of varying concentrations of supernatants with u-PA in a volume of 100 ⁇ l.
  • FIG. 42 Radioimmunoprecipitation of 125 I-labelled purified u-PAR. Ordinate: % 125 I-u-PAR precipitated. Abscissa: dilution of immune/non-immune sera 1:75, 1:750, 1:7500 and 1:75000.
  • Bars 1-11 represent: 1) Total amount of 125 I-u-PAR added to each sample, 44000 cpm; 2) control of binding of radioactivity to the test tubes; 3) control of binding of 125 I-u-PAR to Protein A Sepharose; 4-7) binding of 125 I-u-PAR to non-immune serum; 8-11) binding of 125 I-u-PAR to immune serum.
  • FIG. 43 Reverse solid phase radioimmunoassay, as described under Methods. Catching of 125 I-u-PAR by immune/non-immune sera. Ordinate: cpm bound. Abscissa: a 2-fold serial dilution of antibodies, 1:500-1:32000. (x—x) immune; ( ⁇ — ⁇ ) non-immune. Total amount of 125 I-u-PAR added to each sample: 33000 cpm.
  • FIG. 44 ELISA. Purified u-PAR was coated in a concentration of 1 ng/well. Immune/non-immune sera (primary antibody) were added in a 2-fold serial dilution, ranging from 1:500 to 1:256000. Peroxidase-conjugated secondary antibody diluted 1:500 was used. The substrate was OPD. Colour development from the enzyme substrate reaction was read at 490 nm. The reaction was stopped after 10 minutes. y-axis: OD 490 nm. x-axis: Dilution of immune/non-immune sera. x—x immune; ⁇ — ⁇ non-immune.
  • FIGS. 45A-45B Inhibition of cellular ATF binding by antibodies raised against purified u-PAR. 5 ⁇ 10 5 U937a cells were preincubated with mouse antiserum raised against purified u-PAR ( ⁇ — ⁇ ) or with a control mouse antiserum raised against porcine mucins ( ⁇ — ⁇ ) for 1 hour at 4° C., followed by addition of 2.2 nM 125 I-ATF and incubation for another hour at the same temperature. The cells were then washed 3 times after which the cell-bound radioactivity was measured in a gamma counter.
  • the abscissa represents a two-fold dilution series of the antisera, the final dilutions ranging from 1:153,600 to 1:300.
  • the ordinate axis expresses the cell-associated radioactivity as a percentage of the value obtained with no antiserum present. Substitution of the antiserum with 700 nM unlabelled u-PA led to a 90% inhibition of binding.
  • FIG. 45 B Western blot showing the reactivity of the antisera used.
  • 500 ng of purified u-PAR (lanes 2 and 4) or the Triton X-114 detergent phase obtained from 2.5 ⁇ 10 6 PMA-stimulated U937 cells (lanes 1 and 3) were analyzed by SDS-PAGE under reducing conditions on a 6-16% gradient gel, and Western blotting using as the primary antisera mouse anti-u-PAR serum diluted 1:250 (lanes 1 and 2) or the above control serum at the same dilution (lanes 3 and 4).
  • FIGS. 46A-B show a Western blot, demonstrating the reactivity of polyclonal rabbit antibody against u-PAR.
  • 75 ⁇ l samples of Triton X-114 detergent phase from lysates of PMA-stimulated U937 cells were analyzed alone (lane 1), after mixing with DFP-treated u-PA (Example 1; final concentration 10 ⁇ g/ml) (lane 4), or after mixing with the same amount of DFP-treated u-PA, followed by chemical cross-linking (lane 3).
  • the same amount of DFP-treated u-PA was analyzed alone, after the performance of cross-linking (lane 5), or directly (lane 6).
  • the sample in lane 2 contained 75 ⁇ l of the cell lysate detergent phase, which was subjected to chemical cross-linking without the addition of DFP-treated u-PA.
  • the samples were run on 6-16% gradient SDS-PAGE under non-reducing conditions, followed by electroblotting onto nitrocellulose.
  • the sheets were incubated with purified and absorbed IgG from rabbit anti-u-PAR serum (FIG. 46 A), or with purified and absorbed IgG from pre-immune serum from the same rabbit (FIG. 46 B). The IgG concentration during the incubation was 12 ⁇ g/ml in both cases.
  • the sheets were developed with alkaline phosphatase-coupled antibody against rabbit IgG, followed by detection of alkaline phosphatase activity.
  • FIGS. 47A and B Visualization of the u-PA receptor on the surface of PMA-treated U937 cells. After removal of bound u-PA by acid, cells were incubated either with biotinylated DFP-inactivated u-PA (FIG. 47A) or with biotinylated DFP-inactivated u-PA together with a surplus of unlabelled u-PA (FIG. 47 B). u-PA bound to the cells was detected using FITC-labelled streptavidin.
  • FIGS. 48A and B Visualization of the u-PA receptor in cryostat sections of human chorion. After removal of bound u-PA by acid, the sections were incubated either with biotinylated DFP-inactivated u-PA (FIG. 48A) or with biotinylated DFP-inactivated u-PA together with a surplus of unlabelled u-PA (FIG. 48 B). u-PA bound to the cells was detected using FITC-labelled streptavidin.
  • SDS-PAGE SDS-PAGE.
  • SDS-PAGE was performed according to Laemmli, U.K., “Cleavage of structural proteins during the assembly of the head of bacteriophage T4”, Nature 227: 680-682, 1970, using 6-16% gradient slab gels. Pretreatment of samples under nonreducing conditions was performed without boiling. When reducing conditions were used, the samples were boiled for 5 minutes in the presence of 20 mM DTT.
  • PHASTGELTM gel electrophoresis SDS-PAGE was performed on a Phast gel apparatus (Pharmacia), using ready-made 10-15% gradient gels. Electrophoresis was performed according to the recommendations of the manufacturer. Silver staining was performed according to Heukeshoven and Dernick, 1988.
  • Tricine-SDS-PAGE of samples to be electroblotted for amino acid analysis or NH 2 -terminal amino-acid sequencing was performed in a MINI-PROTEAN IITM amino acid sequencing apparatus (BioRad) according to Shugger and von Jagow, 1987, on a 0.75 mm homogeneous 7.7% T, 3% C gel.
  • the gel was pre-electrophoresed for 3 hours at 15 mA in the gel buffer with 12 mM 3-mercaptopropanoic acid added as a scavenger.
  • the freeze-dried sample was dissolved directly in 50 ⁇ l of the sample buffer with 40 mM dithioerythritol as the reducing agent, and boiled for 2 minutes.
  • the gel buffer used for pre-electrophoresis was replaced with electrophoresis buffer, after which electrophoresis was performed for 4 hours at 60 V.
  • Electroblotting of samples for amino acid analysis or NH 2 -terminal amino acid sequencing was electroblotted onto a polyvinylidene difluoride (PVDF) membrane (Millipore), using a semi-dry electroblotting apparatus (JKA Instruments, Denmark). Electroblotting took place at pH 11.0 in 10 mM CAPS (3-(cyclohexylamino)-1-propanesulfonic acid), including 0.4 mM dithioerythritol and 10% methanol, and was performed at 0.8 mA/cm 2 for 2 hours. The protein was localized by staining with Coomassie R250 for 2 minutes and brief destaining, followed by wash in water (Matsudaira, 1987).
  • PVDF polyvinylidene difluoride
  • the Coomassie-stained protein band was cut out from the PVDF-membrane and treated with 25 mM iodoacetamide in 50 mM sodium borate, pH 8.0, for 1 hour in the dark at room temperature. After the reaction, it was washed extensively with water and dried under argon. The protein on the dried filter was sequenced on an Applied Biosystems protein sequencer, model 477A.
  • the on-line HPLC identification system for the PTH amino acid derivatives included the derivative of carboxymethylcysteine (produced by deamidation of the amidomethyl derivative during conversion). The correct identification of this derivative was assured by a test-sequencing of chicken lysozyme (with cysteine at residue no. 6) after parallel preparative electrophoresis, electroblotting and alkylation.
  • U937a histiocytic lymphoma cell line U937 (here designated as U937a) (E.K.O. Kruithof, University Hospital Center, Lausanne, Switzerland), a variant of this cell line, designated U937b (A. Fattorossi, Research Lab of Aeronautica Militare, Rome, Italy), the promyeloid leukemic cell line HL-60 (American Type Culture Collection (ATCC)), the bladder carcinoma cell line 5637 (ATCC), the larynx epidermoid carcinoma cell line HEp-2 (ATCC), the epidermoid carcinoma cell line A-431 (E.
  • the cervix carcinoma cell line HeLa (ATCC), the colon carcinoma cell line HCT 116 (ATCC), the conjunctiva cell line Chang (ATCC), the choriocarcinoma cell line JEG-3 (A. Vaheri, University of Helsinki, Finland), the amnion cell line AV3 (ATCC), and the fibrosarcoma cell line HT-1080 (A. Vaheri).
  • the U937a and b and HL-60 cells were grown in suspension, while all the other cell lines were grown as monolayers.
  • the HT-1080 and A-431 cells were grown in Dulbecco's modified Eagle's medium with 10% heat-inactivated fetal calf serum.
  • U937a cells Large-scale production of U937a cells.
  • the U937a cells were grown in 1-liter spinner flasks to reach a density of 1.0-1.5 ⁇ 10 6 cells/ml in RPMI 1640 medium supplemented with 2 mM L-glutamine, 5% fetal calf serum (heat inactivated), 200 units/ml penicillin, 25 ⁇ g/ml streptomycin (or without antibiotics). Each flask contained 500 ml cell culture.
  • Phorbol 12-myristate 13-acetate (PMA) induction and harvest of U937a cells The 500 ml cell suspension of one spinner flask was added to 1 liter of fresh medium without serum. 150 ⁇ l of PMA stock solution in dimethylsulfoxide (1 mg PMA/ml) was added, to reach a final concentration of 150 nM PMA. The culture was transferred to a 10-layer cell factory (Nunc, Denmark) and grown for 3.5 days in the factory. Upon addition of the PMA solution, the cells stop dividing and attach to the surface.
  • the clarified lysate was subjected to temperature-induced phase separation (Bordier, 1981) by incubation at 37° C. for 10 minutes, after which the detergent phase was collected by centrifugation for 10 minutes at 20° C., 1,800 ⁇ g. The upper phase was discarded.
  • the lower phase (approximately 2 ml) was washed by addition of 18 ml 0.1 M Tris/HCl, pH 8.1, at 0° C., followed by complete mixing to restore a clear, one-phase solution, and repeated phase separation by warming and centrifugation, as above.
  • the lower phase was made up to 20 ml by addition of 0.1 M Tris/HCl, pH 8.1.
  • 500 ⁇ l 10% w/v 3-((3-cholamidopropyl)dimethylammonio)-1-propanesulfonate (CHAPS) was added to yield a clear, single-phase detergent fraction. Minor amounts of non-dissolved material were removed from this solution by centrifugation for 15 minutes at 4° C., 3,300 ⁇ g.
  • Lysates and detergent phases from other cell types were prepared in the same manner, except that smaller amounts of cell material were used. The amounts of all reagents were reduced proportionally. In one experiment, 0.5% CHAPS was used as the lysis detergent instead of 1% Triton X114. In that experiment, no phase separation was performed.
  • the reaction was stopped by extensive dialysis at 0° C. against 0.25 M NaHCO 3 , 0.5 M NaCl, 0.1% Triton X-100, pH 8.5.
  • the dialyzed material was coupled to 12.5 ml of CNBr-activated Sepharose (Pharmacia) that had been freshly equilibrated with 0.25 M NaHCO 3 , 0.5 M NaCl, pH 8.5 (coupling buffer).
  • the reaction proceeded overnight at 4° C. and was stopped by equilibration of the matrix with 1 M ethanolamine/HCl, pH 8.0 and incubation for 24 hours at 4° C.
  • the matrix (DFP-u-PA-Sepharose) was washed with the coupling buffer and pre-eluted with the appropriate elution buffer (see below) before use.
  • the clarified detergent fraction obtained from 6 ⁇ 10 9 U937a cells was diluted with 1 vol washing buffer-1 (10 mM sodium phosphate, 140 mM sodium chloride, 0.1% CHAPS, pH 7.4) and chromatographed on a column containing 8 ml DFP-u-PA-Sepharose, equilibrated with the same buffer. After application of the sample, the column was washed with washing buffer-1, followed by washing buffer-2 (10 mM sodium phosphate, 1 M sodium chloride, 0.1% CHAPS, pH 7.4). The column was eluted from below with elution buffer (0.1 M acetic acid, 0.5 M sodium chloride, 0.1% CHAPS, pH 2.5).
  • Elution fractions were immediately titrated to pH 7.5 by addition of the appropriate volume of 0.1 M sodium phosphate, 1.0 M sodium carbonate, pH 9.0.
  • u-PAR-containing fractions were identified by chemical cross-linking to the 125 I-labelled amino terminal (ATF) fragment of urokinase, followed by SDS-PAGE and autoradiography.
  • Purified u-PAR samples for amino acid analysis or NH 2 -terminal amino acid sequencing were dialyzed against 0.1% acetic acid and lyophilized.
  • Protein labelling with 125 I was performed as described previously (Nielsen et al., 1988), except that 0.1% Triton X100 was replaced by 0.01% Tween 80. Purified u-PAR, concentrated by freeze-drying after dialysis against 0.1% acetic acid, was iodinated in the same manner, except that 1.5 ⁇ g protein was treated with 250 ⁇ Ci 125 I in a volume of 25 ⁇ l.
  • Lyophilized, purified u-PAR was radioiodinated directly.
  • the samples were denatured under mildly reducing conditions by the addition of SDS and dithiothreitol to final concentrations of 0.5% and 1.6 mM, respectively, and boiling for 3 minutes. Aliquots of the denatured samples (10 ⁇ l) were adjusted to include 200 mM sodium phosphate, pH 8.6, 1.5% Triton X-100, 10 mM 1,10 phenanthroline (added from a methanol stock solution) and either 1 unit of peptide:N-glycosidase F (N-glycanase, Genzyme), or no enzyme, in a total volume 30 ⁇ l. Deglycosylation was performed at 37° C. for 20 hours.
  • 70 ⁇ l lysate samples labelled by cross-linking to 125 I-ATF were made up to 200 ⁇ l with 0.05 M sodium acetate, pH 5.0. 90 ⁇ l aliquots of the mixture received either 14 ⁇ l of 33 ng/ ⁇ l neuraminidase (Boehringer-Mannheim) or no enzyme. Desialylation was performed overnight at 37° C.
  • PMA-stimulated U937a cells were acid-treated to remove any surface-bound u-PA and lysed in a Triton X114 containing buffer.
  • the detergent extract was subjected to temperature-induced phase separation, and the isolated detergent phase was used as the raw material for affinity chromatography.
  • the acid eluates were neutralized and analyzed, either directly or after concentration by dialysis against 0.1% acetic acid and lyophilization.
  • the electrophoretic appearance of the purified material is shown in FIGS. 1A-C.
  • FIG. 1 A After SDS-PAGE and silver staining (FIG. 1 A), the eluted protein migrated as one broad band, covering the range from approximately 55 to 60 kDa. Outside this range, no protein material was detected. A single band with the same apparent molecular mass was also found when SDS-PAGE was performed under nonreducing conditions. (FIG. 1C, lane 5).
  • Binding and cross-linking to 125 I-labelled ATF was specific and saturable. Thus, it could be competed for by an excess of unlabelled ATF, active u-PA or DFP-treated u-PA, while no competition was obtained with unrelated proteins such as, for example, bovine serum albumin, or with related proteins, such as t-PA, plasminogen or epidermal growth factor (FIG. 1 B).
  • unrelated proteins such as, for example, bovine serum albumin, or with related proteins, such as t-PA, plasminogen or epidermal growth factor (FIG. 1 B).
  • Quantification by amino acid analysis indicated a purification yield of 6-9 ⁇ g polypeptide (corresponding to about 10-15 ⁇ g u-PAR glycoprotein; see below) from 6 ⁇ 10 9 cells.
  • Amino acid composition and NH 2 -terminal amino acid sequences are shown in table 1.
  • This composition includes a strikingly high content of cysteine residues. Further, it is noted that rather few lysine residues are present.
  • the analysis system employed allows the quantification of glucosamine and galactosamine in addition to the amino acids. Glucosamine was detected in an amount corresponding to approximately 30 mol of N-acetylglucosamine per mol protein, correcting for loss during hydrolysis. In contrast, no galactosamine was identified.
  • the amino terminus like the amino acid composition of the entire protein, is rich in cysteine residues.
  • Example 2 Data for probe construction (Example 2) were derived from the sequencing shown in Table 2A. For this construction, position 6 of the amino acid sequence was tentatively assigned Asn; see footnote a of Table 2A.
  • FIG. 2 shows the electrophoretic appearance of he deglycosylated protein. The electrophoretic band observed after autoradiography of the 125 I-labelled protein was always slightly broader than that seen after direct protein staining.
  • the reaction turned the heterogeneous 55-60 kDa receptor (lane 1) into a deglycosylated protein of only 35 kDa that migrated as a much sharper band (lane 2), thus further confirming that the initially heterogeneous material all represented variants of the same protein.
  • the fibrosarcoma cell line HT-1080 contained a third u-PAR variant, giving rise to a single conjugate band of a slightly lower molecular weight (approximately 65 kDa; not shown).
  • N-bound glycosylation was responsible, not only for molecular u-PAR heterogeneity within the PMA-stimulated U937 line and occurrence of two bands in the PMA-stimulated U937b line, but also for the electrophoretic difference between u-PARs from non-stimulated and PMA-stimulated U937 cells and for the variation among different cell lines (i.e. HT-1080 fibrosarcoma cells compared to the other cell lines tested).
  • Enzymatic degradation Affinity purified u-PAR was dialyzed against 0.1% acetic acid and lyophilized as described in Example 1. The freeze-dried material was redissolved in incubation buffer (0.05 M Tris/HCl, 0.05% CHAPS, pH 8.1) to yield a protein concentration of approx 25 ⁇ g/ml. 9 ⁇ l samples of this u-PAR solution were treated with chymotrypsin (Worthington; final concentrations ranging from 8-200 ng/ml), by addition of 1 ⁇ l of the appropriate stock solution of the enzyme, dissolved in incubation buffer. The samples were incubated for 16 h at 37° C. after which the degradation was stopped by addition of 0.5 ⁇ l of 20 mM phenylmethylsulfonylfluoride, dissolved in dimethylsulfoxide. The samples were stored at ⁇ 80° C. until analysis.
  • incubation buffer 0.05 M Tris/HCl, 0.05% CHAPS, pH 8.1
  • Samples to be analyzed by chemical cross-linking to 125 I-ATF were 50-fold diluted in 0.1 M Tris/HCl, 1% Triton X-114, pH 8.1.
  • the diluted samples were either clarified by addition of 0.25% w/v CHAPS (final concentration) or subjected to a single round of temperature induced phase separation (see Example 1).
  • each phase i.e., the detergent and buffer fraction, respectively
  • Enzymatic deglycosylation with N-Glycanase was performed according to example 1, except that the actual concentrations during the deglycosylation step were the following: 0.08% SDS; 0.26 mM dithiothreitol; 0.11 M sodium phosphate; 0.9% Triton X-100; 5.3 mM 1,10 phenanthroline; 33.3 units/ml N-glycanase.
  • Identification of the fragment will subsequently be done by comparison to the amino acid sequence derived from u-PAR cDNA.
  • synthetic peptides covering the chymotryptic fragment will be constructed. The peptides will be assayed for their potential inhibitory activity against the binding reaction between u-PAR and the ligand, as studied by cell binding assays (Nielsen et al., 1988; Appella et al., 1987) or by chemical cross-linking assay.
  • the samples were analyzed in the chemical cross-linking assay, using 125 I-ATF as the ligand (FIG. 5 ). While the non-degraded samples (lanes 4 and 5) showed the 70-75 kD conjugate band which is characteristic for the intact u-PAR (see Example 1), the intensity of this band was much reduced in the degraded samples (lanes 1-3). In contrast, the degraded samples showed an approx. ⁇ 30 kD cross-linked conjugate; i.e. the size to be expected for a conjugate formed between the above mentioned, 16 kD u-PAR degradation product and the 15 kD ATF.
  • the ligand binding fragment proved hydrophilic in the Triton X-114 system, suggesting that this fragment does not include the diacylglycerol part of the protein (see Example 4).
  • the deglycosylation experiment showed that the ligand binding fragment is glycosylated and suggested that the polypeptide part of the fragment comprised only 6-10 kD, corresponding to approx. 50-90 amino acid residues.
  • a human cDNA library was used made from SV 40 transformed human GM637 fibroblasts in a plasmid vector based on pBR322 (carrying an ampicillin resistance gene) (Okayama H, Berg P, “High-efficiency cloning of full-length cDNA”, Mol. Cell. Biol. 2: 161-170, 1982). The library was kindly donated by Dr. Okayama. This library was selected on the basis of the known high number of u-PAR in GM637 cells (Blasi, unpublished).
  • the plasmid vector (FIG. 8) uses the SV 40 promoter and has high expression in various eukaryotic cells, but very low or no expression in prokaryotes.
  • the library was screened with synthetic oligonucleotide probes made on the basis of amino acid sequence data from purified receptor protein (Tables 4-5).
  • the melting temperatures were calculated from Lathe, J. Mol. Biol. 183: 1-12, 1985.
  • the equation used was modified from:
  • l is the length of the DNA (number of bases), and h is the percent homology.
  • nitrocellulose filters containing DNA from the plasmid library were hybridized to the end-labelled oligonucleotide probe at various temperatures and salt concentrations (all within the range calculated from Lathe, supra).
  • the filters were produced according to Grunstein and Hogness (“Colony hybridization: A method for the isolation of cloned DNAs that contain a specific gene”, Proc. Natl. Acad. Sci. USA 72: 3961, 1975).
  • the hybridization conditions to be used for the screening were chosen as the ones giving the minimum amount of background hybridization.
  • Table 3 the amino acid sequence derived frm a preliminary amino-terminal sequencing of purified u-PAR (see Example 1) and the derived oligonucleotide sequence are presented.
  • the plasmid library was screened with the N-terminal probe using the procedure of Crunstein and Hogness (supra). The detailed procedure is described below. Several positive clones were found but after the third rescreening, only one remained. The purity of the clone was checked and DNA was prepared from it (see large scale DNA preparation below). The DNA was digested with several different restrictions enzymes, and a map of the restriction sites found in the clone was constructed (see procedure in Maniatis et al., Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory, 1982). The insert was further analysed by DNA sequencing (see procedure below).
  • the clone was able to code for 7 out of the 8 amino acids in the N-terminal peptide used to construct the 24-mer probe.
  • the sequence in the probe starts with an A whereas the clone had a T in this position, resulting in the substitution of Cys for Met.
  • the clone was thus isolated by a specific hybridization but could not code for the correct peptide.
  • the library was then rescreened using chloramphenicol amplification of plasmids (Maniatis et al., supra) to increase the signal intensity. This procedure resulted in a total of 7 positive clones (see Table 4) of which two were eliminated on the basis of hybridization to the probe made from the original false positive clone.
  • This primer was made in the inventors' laboratory on an Applied Biosystems 391 A DNA Synthesizer. Primers for the pEMBL18 vector were purchased from Biolabs.
  • Plasmid DNA was prepared according to described procedures (Maniatis, supra).
  • the synthetic oligonucleotides were end-labelled using T4 polynucleotide kinase and ⁇ - 32 P-ATP.
  • Gel purified DNA fragments were nick-translated using a BRL nick translation kit and ⁇ - 32 P-ATP.
  • the probes were purified on NENSORB 20 columns (NEN) following the manufacturer's specifications. Nick-translated probes were denatured for 10 minutes at 100° C. before use.
  • the complete sequence of one of the isolated clones was obtained on double-stranded DNA in both orientations using commercial primers for pEMBL18 (M13 primers) and internal synthetic primers (see above).
  • the sequence is shown in Sequence (1) in the Detailed Description of the invention.
  • the restriction map and the sequencing strategy are illustrated in FIGS. 7A-C.
  • the cDNA clone is 1364 nucleotides long from the 5′ end to the beginning of the polyA stretch. At the 5′ end, 46 nucleotides precede the first ATG codon which is followed by a 1005 nucleotides sequence with an open reading frame, ending with a nonanucleotide containing two in frame stop codons.
  • 312 nucleotides of 3′ untranslated sequence separate the first stop (TAA) codon from the polyA sequence.
  • TAA first stop codon
  • the assignment of the ATG at nucleotide 47 as the translation start site agrees with the consensus for initiating regions (Kozak, 1987) as discussed above.
  • the translated sequence starts with a hydrophobic sequence which conforms to the rules for the signal peptide (von Heijne, 1986) (see above).
  • the putative signal peptide is followed by 313 amino acid residues.
  • the sequence shown in Sequence (1) was compared with the initial amino terminal sequence (FIG. 7 A), and it was observed that in fact the original sequence contained an error at position 6 (Asn instead of Cys) which, however, did not prevent the isolation of the right cDNA clone.
  • the human u-PAR is a relatively small protein of 313 amino acid residues.
  • the amino acid sequence contains five potential N-linked glycosylation sites, in agreement with the high level of glycosylation of the protein (see Example 1).
  • a sequence of 21 hydrophobic amino acids flanked by arginine residues may represent a membrane spanning domain of the u-PAR (FIG. 7 C).
  • the arginine is followed by 9 additional hydrophobic amino acids ending with a carboxy-terminal threonine.
  • u-PAR may contain no intracytoplasmic domain at all, i.e. also the carboxy-terminal 10 residues may be buried in the membrane.
  • the sequence of the carboxy-terminal about 30 amino acid residues would also be compatible with a signal peptide for glycolipid-anchored, phospholipase C-sensitive membrane attachment (Ferguson and Williams, 1988).
  • the u-PAR is a slightly acidic protein (6 net acid charges), is very rich in cysteine, rich in glycine and leucine, and poor in lysine.
  • the u-PAR is also rich in serine and threonine residues, which might indicate O-linked glycosylation (Russell et al., 1984). However, deglycosylation and sugar composition studies indicate that the receptor contains only N-linked carbohydrates (see Example 1).
  • the u-PAR sequence is not similar to any known protein: a search in the Georgetown University data bank did not yield any extended homology. In particular, it bears no resemblance to the tissue factor, a receptor for factor VII of the coagulation pathway, which in common with u-PAR has the low molecular weight and the unusually large extent of glycosylation (Morrissey et al., 1987).
  • the very high proportion of cysteine residues is common to many extracellular portions of receptors, like the epidermal growth factor receptor (Yarden & Ullrich, 1988), the epidermal growth factor precursor (Bell et al., 1986), and many others (Appella et al., 1988). However, there does not appear to be a common pattern of cysteine spacings in these proteins.
  • the second and third repeats are the most closely related (about 25 percent identity). Significantly, the pattern of cysteines is strikingly similar in these two repeats.
  • p-u-PAR-1 clone The functionality of p-u-PAR-1 clone was tested by transfecting it into mouse LB6 cells and testing transfectants by the caseinolytic plaque assay. This assay is based on the ability of plasmin to degrade casein which gives rise to clear plaques in an opaque background. Since LB6 cells produce no plasminogen activator, plasmin cannot be produced. In the presence of u-PA receptors, however, cells can bind u-PA and hence acquire the ability to degrade casein (Vassalli et al., 1985). The murine LB6 cells produce no plasminogen activator (unpublished observation) but have u-PA receptors.
  • LB6 cells cannot bind human u-PA.
  • Expression of human u-PAR cDNA by LB6 cells should provide these cells with the ability to bind human u-PA which can be visualized by the formation of clear plaques in the caseinolytic plaque assay.
  • the vector used in the cDNA library is an expression vector that contains the SV40 promoter at the 5′ end and polyadenylation and splice sites at the 3′ end (Okayama & Berg, 1983). Expression of human u-PA receptors in transfected cells will, therefore, prove that the p-u-PAR-1 clone encodes a complete cDNA sequence.
  • Mouse LB6 cells (Corsaro and Pearson, 1981) were cultured in Dulbecco's modified minimal essential medium (DMEM) supplemented with 10% foetal calf serum, 2 mM glutamine and 10 IU/ml of penicillin and streptomycin.
  • Human high molecular weight urokinase and prourokinase were provided by Lepetit SpA (Nolli et al., 1989).
  • the amino terminal fragment of human u-PA, ATF was a gift from Abbott Laboratories.
  • the synthetic peptides human u-PA[12-32(ala19)] and mouse u-PA[13-33(ala20)] have been described before (Appella et al., 1987). Plasminogen was from Sigma Chemical Co.
  • 2 ⁇ 10 5 LB6 cells were transfected either with 9 ⁇ g of p-u-PAR-1 DNA plus 1 ⁇ g of pRSVneo DNA, or with 9 ⁇ g of pRSVCAT plous 1 ⁇ g of pRSVneo DNA using a modification of the calcium phosphate coprecipitation technique (Pozzatti et al., 1986). Cells were plated in 0.8 mg/ml G418-containing DMEM, 10% foetal calf serum, and colonies were isolated after about 13 days.
  • the pools of transfected clones were tested (in the case of p-u-PAR-1 DNA) by the caseinolytic plaque assay (Vassalli et al., 1977) and positive clones were picked. After one subcloning, several clones from each transfection were tested for human u-PA binding using the same technique.
  • Cells (plated one day before at 100,000/dish) were washed with PBS, incubated in the presence of 0.2 nM human u-PA for 1 hour at 37° C., washed extensively and covered with a thin agar layer containing 1.3% casein and 17 ⁇ g/ml plasminogen. The plates were incubated at 37° C. for 3 hours, stained with Coomassie brilliant blue R and photographed. In some experiments, specific competitors were used during the binding step.
  • p-u-PAR-1 and pRSVneo DNA was cotransfected into mouse LB6 cells and a pool of G418 resistant clones was isolated and analysed for human u-PA binding (0.2 nM) by the caseinolytic plaque technique. Control experiments showed that all cells were negative in this assay in the absence of added u-PA or plasminogen. After incubation with human u-PA and in the presence of plasminogen, the pool of G418 resistant cells that had received p-u-PAR-1 DNA gave a high number of caseinolytic plaques; control cells (transfected with pRSVCAT and pRSVneo DNA) were negative (data not shown).
  • FIGS. 9A-F LB6 cells transfected with p-u-PAR-1 DNA formed caseinolytic plaques upon binding human u-PA (see FIGS. 9A vs. 9 B), whereas those transfected with pRSVCAT DNA did not (see FIG. 9 B).
  • ATF amino-terminal fragment of u-PA
  • FIG. 9B Specificity is shown by the ability of the amino-terminal fragment of u-PA (ATF), i.e. a truncated u-PA molecule maintaining the binding capacity but deprived of the catalytic activity (Stoppelli et al., 1985) (FIG.
  • the expression of the human u-PAR by mouse LB6 cells transfected with p-u-PAR-1 was further analysed by binding competition experiments using unlabelled and iodinated ATF.
  • the molecular properties of the u-PAR expressed by the transfected cells were analysed by SDS-PACE and radiography of material from these cells cross-linked to iodinated ATF.
  • Mouse LB6 cells were grown in DMEM as described in this Example. Iodination of ATF has been described previously by Stoppelli et al. (1985). The cross-linking reagent disuccinimidyl suberate was from Pierce Chemical Co.
  • LB6/RSVCAT or LB6/p-u-PAR-1 cells in a 30 mm dish were washed with PBS containing 1 mg/ml bovine serum albumin, incubated in serum-free medium for 1 hour at 37° C., and then incubated with 47,000 cpm 125 I-ATF (1500 cpm/fmole) at 37° C. for 60 minutes in the presence of different concentrations of unlabelled ATF.
  • the experiment was carried out in duplicate.
  • the cells were washed with PBS-bovine serum albumin, incubated for 15 minutes at 37° C. in 0.5 N NaOH, and the cell lysate was collected and counted (Stoppelli et al., 1985). Specific binding was calculated by subtracting the radioactivity not competed by 100 nM ATF.
  • Cross-linking was stopped with 10 mM (final concentration) ammonium acetate and incubated for 10 minutes at room temperature.
  • Cells were scraped with PBS containing 1 mM EDTA, 1 mM PMSF, collected by centrifugation, resuspended in 25 ⁇ l of distilled water, and counted. The cells were then lysed directly in Laemmli buffer containing 5% ⁇ -mercaptoethanol (Laemmli, 1970). In control samples, 100 nM unlabelled ATF was present during the binding step.
  • the cell extract was analysed by SDS-polyacrylamide (12.5%) gel electrophoresis under reducing conditions (Laemmli, 1970), along with molecular weight markers (Rainbow, Amersham) (myosin; phosphorylase b; bovine serum albumin; ovalbumin; carbonic anhydrase; trypsin inhibitor; lysozyme).
  • SDS-polyacrylamide (12.5%) gel electrophoresis under reducing conditions (Laemmli, 1970), along with molecular weight markers (Rainbow, Amersham) (myosin; phosphorylase b; bovine serum albumin; ovalbumin; carbonic anhydrase; trypsin inhibitor; lysozyme).
  • the gel was dried and exposed to X-ray film.
  • FIG. 10A shows a binding-competition plot in which control LB6 cells (LBS/RSVCAT) do not bind 125 I-ATF, whereas LB6 cells transfected with p-u-PAR-1 DNA do. The binding is specifically competed by unlabelled ATF. Scatchard plot of the data gave a Ka of about 10 8 moles ⁇ 1 and about 25,000 receptors/cell.
  • migration of the cross-linked ligand corresponds to a molecular weight of slightly less than 69,000 identical to that obtained with human GM637 cells (from which the cDNA clone is derived). This is the molecular weight expected for the intact ATF-u-PAR complex (Nielsen et al., 1988).
  • the data presented in FIG. 10B are in perfect agreement with those obtained with purified u-PAR (Nielsen et al., 1988).
  • This Example shows expression of the human u-PAR gene in mouse LB6 cells by the following findings: p-u-PAR-1 DNA transfected LB6 cells bind labelled human ATF and unlabelled human u-PA as shown by direct binding assay (FIG. 10A) and the caseinolytic plaque assay (FIG. 9 ). The binding is specific as shown by the ability of human ATF, human synthetic peptide u-PA[12-32(ala19)], but not mouse synthetic peptide u-PA[13-33(ala20)] to compete for binding (FIGS. 9A-F and 10 A). The ATF-u-PAR complex has the correct molecular weight (FIG. 10 B).
  • Receptors are anchored at the plasma membrane by a stretch of hydrophobic amino acids (the trans-membrane domain) or through a glycolipid anchor.
  • Most integral membrane proteins have a single trans-membrane domain, although cases have been described of multiple trans-membrane domains.
  • the trans-membrane domain is present in the middle of the protein sequence, i.e. between the carboxy terminal portion (generally intracellular) and the amino terminus (generally extracellular, containing the binding site for the ligand in the case of most receptors).
  • a carboxy-terminal hydrophobic region is also a signal for glycolipid-anchor processing.
  • the available information on the structure of the u-PAR indicates that it is a protein of about 35,000 daltons, i.e. about 330 amino acids.
  • the sequence cut by the restriction endonuclease PFLM-1 is:
  • p-u-PAR-1 DNA was cut with PFLM-1, the ends filled with T4 DNA polymerase to produce
  • p-u-PAR-PFLM-1 clone has been transfected into LB6 cells as described above and its expression compared with that of wildtype p-u-PAR-1 cDNA.
  • this mutant expresses a u-PAR molecule that is partly recovered in the medium and partly retained in the cells.
  • cross-linking to iodinated ATF shows a single band in the medium and two bands in the Triton X-114 extract (prepared as described in Example 1).
  • the lower molecular weight band corresponds to a molecular weight of the non-glycosylated u-PAR. Only the high molecular weight band is present on the cell surface (see below).
  • the data presented in FIG. 11 indicate that approximately 10 times as much protein is present in the medium with respect to what is retained in the cell.
  • a second mutant has been prepared in which the carboxy-terminal 36 amino acids have been deleted from the u-PA receptor, thus leaving a protein with no trans-membrane and no glycolipid anchor domain.
  • oligonucleotide-directed mutagenesis was employed, using the system commercially available from Amersham, to insert a single EcoRV site. To this end, the following oligonucleotide was used which hybridizes to the nucleotides 935-952 of the u-PAR cDNA sequence:
  • This mutant (p-uPAR-278stop) is expected to be unable to attach to the cell surface, to be secreted in the medium, and to bind pro-u-PA, ATF, DFP-u-PA and active u-PA, in general the same molecules bound by the normal u-PA receptor. It should therefore be useful as a u-PA or pro-u-PA scavenger in all the cases where a reduction of u-PA activity is desired.
  • u-PAR has a Glycosyl-Phosphotidylinositol Anchor and is C-Terminally Processed
  • PVDF membranes (Immobilon-P) were from Millipore. N w ,N ′w -dimethyl-Arg was from Sigma, N w -monomethyl-Arg from Calbiochem, whereas N w ,N w -dimethyl-Arg was a kind gift from Dr. T. Ogawa (University of Tokushima, Japan). Ethanolamine was from Merck. Na 125 I, [9,10(n)- 3 H]-myristic acid (53 Ci/mmol), myo-[2- 3 H]inositol (18.3 Ci/mmol) and [1- 3 H]ethanolamine hydrochloride (19 Ci/mmol) were from Amersham.
  • Acetylcholinesterases from human and bovine erythrocytes, phospholipase A 2 from bee venom and myelin basic protein from bovine brain were from Sigma.
  • Phospholipase D from cabbage and phosphatidylinositol-specific phospholipase C from Bacillus cereus (PI-PLC) were from Boehringer Mannheim.
  • u-PAR was purified from PMA-stimulated U937 cells as in Example 1. Active human u-PA was purchaged from Serono and was DFP-inactivated as described (Nielsen et al., 1988); the amino terminal fragment (ATF) of u-PA was a kind gift from Dr. G.
  • ATF, u-PAR and DFP-inhibited u-PA were radio-labelled as described (Nielsen et al., 1988) except that 0.1% (v/v) Triton X-100 was replaced by 0.1% (w/v) CHAPS in the case of u-PAR and by 0.01% (v/v) Tween 80 in the case of ATF and DFP-u-PA.
  • Preparation of polyclonal rabbit antibodies against human u-PAR was carried out as described in Example 11.
  • exogenously added 125 I-labelled DFP-uPA (1 nM) were allowed to rebind to the unoccupied u-PAR by incubation for 2 hours at 4° C. in buffer A followed by 3 ⁇ wash in the same buffer without added ligand. Incubation of these adherent U937 cells with the various phospholipases were performed in buffer A at 37° C. on a shaking table.
  • Both media were supplemented with: 2 mM L-glutamine, 5 mM Na-pyruvate, 200 units/ml penicillin, 25 ⁇ g/ml streptomycin, 25 mM HEPES (pH 7.4), 0.5 mg/ml defatted BSA and 4 ⁇ normal concentration of non-essential amino acids. All tracers were added from stock solutions in 25 mg/ml defatted BSA, 0.1 M HEPES (pH 7.4) to a final concentration of 0.1 mCi/ml in 10 ml media and metabolic labelling was allowed to proceed for 15 hours at 37° C.
  • Tricine-SDS-polyacrylamide gels were prepared according to Schwarz and von Jagow, 1987 in a Bio-Rad Mini-Protean II apparatus (8 cm ⁇ 7 cm ⁇ 0.75 mm). The homogenous gel (7.5% T and 3% C) was cast 1 day in advance and subjected to pre-electrophoresis at pH 8.45 with 0.5 M Tris, 0.1% (w/v) SDS and 12 mM 3-mercaptopropionic acid (added as scavenger) for 4 hours at 15 mA/gel.
  • Purified, lyophilized u-PAR was reduced by boiling for 2 minutes in 4% (w/v) SDS, 12% (w/v) glycerol, 50 mM Tris and 40 mM dithiotreitol at pH 6.8.
  • the gel buffer used for pre-electrophoresis was replaced with the original electrophoresis buffer (Schagger and von Janow, 1987) except that 1 mM 3-mercaptopropionic acid was included in the catode buffer. Electrophoresis was performed at 60 V for 4 hours.
  • Electrotransfer onto a 0.45 ⁇ m PVDF-membrane was performed at pH 11 in 10 mM 3-(cyclohexylamino)-1-propane sulfonic acid, 10% v/v methanol and 0.4 mM dithiotreitol by the semi-dry approach at 0.8 mA/cm 2 for 2 hours as previously described (Ploug et al., 1989).
  • the Coomassie stained u-PAR was prepared for amino acid analysis by acid hydrolysis directly on the excised PVDF-membrane at 110° C. in 100 ⁇ l of redistilled 6M HCl including 0.05% (w/v) phenol and 5 ⁇ l of 1% (w/v) DTDPA in 2 M NaOH as published (Ploug et al., 1989). Amino acid analysis was performed on a Waters amino acid analyzer, equipped with o-phtaldialdehyde derivatization essentially as described (Barkholt and Jensen, 1989). However, the chromatographic system was modified slightly to increase resolution of basic amino acids.
  • Elution was still performed by a pH-gradient resulting from mixing two non-halide buffers A and B (for composition see Barkholt and Jensen, 1989), but the gradient consisted of the following linear segments: initial eluant 100% A, 88% A and 12% B at 15 min, 60% A and 40% B at 24 min, 55% A and 45% B at 26 min, 50% A and 50% B at 36 min, 30% A and 70 B at 40 min, 25% A and 75% B at 64 min, 100% A at 65 min and 100% A from 65 to 70 min.
  • Direct autoradiography 125 I
  • fluorography 3 H
  • X-ray film Kodak X-Omat
  • intensifying screens Cronex
  • the X-ray film was pre-exposed (0.2-0.3 A) and the polyacrylamide gels were impregnated with Amplify according to the manufacturer's instructions (Amersham).
  • Acetylcholinesterases isolated from erythrocytes contain ethanolamine as a covalent constituent in a glycolipid membrane anchor, while myelin basic protein posseses a partly methylated arginine residue. It is therefore concluded that u-PAR does contain ethanolamine, covalently linked to the protein by acid labile bonds (e.g. ester or amide bonds). Quantitative analysis of the data in FIG. 12 shows that each u-PAR molecule contains 2-3 ethanolamine residues (see also Table 5).
  • GPI glycosylphosphatidylinositol
  • PI-PLC bacterial phosphatidylinositol-specific phospholipase C
  • u-PA released to the medium by PI-PLC was essentially non-degraded and consisted primarily of intact two-chain u-PA (Mr 50,000) along with a smaller amount of its amino terminal fragment (ATF, Mr 17,000).
  • the receptor-binding domain of u-PA resides in both of these components (Appella et al., 1987). Accordingly, these two molecular species did bind to the cell surface during preincubation with 125 I-labelled DFP-u-PA. In contrast, the low molecular weight form of u-PA (Mr 33,000), devoid of the receptor-binding domain, was eliminated by the washing procedures.
  • a slow, endogenous release of u-PAR could, however, be detected after prolonged incubation in serum-free media without PI-PLC treatment (FIG. 14 C); this finding may indicate that the cells either produce and secrete a soluble u-PAR or more likely, that they produce a GPI-specific phospholipase.
  • Biosynthetic labelling of a component capable of binding to DFP-u-PA, was obtained after incubation of PMA-stimulated U937 cells with either [ 3 H]-ethanolanine, myo-[ 3 H]-inositol or [ 3 H]-myristic acid (data not shown).
  • This protein was isolated from the detergent lysates of U937 cells by immunoprecipitation with specific polyclonal antibodies against u-PAR and analysed by SDS-PAGE and fluorography (see Materials and Methods).
  • f SD standard deviation (absolute number of residues).
  • Phorbol 12-myristate 13-acetate (PMA), dexamethasone and dibutyl cyclic AMP were obtained from Sigma.
  • Deoxycytidine 5′-[ ⁇ - 32 P] triphosphate (specific activity 3000 Ci/mmol), and Rainbow [ 14 C] protein molecular weight markers were purchased from The Radiochemical Centre, Amersham, U.K.
  • a kit for random primed labelling reaction and murine epidermal growth factor (mEGF) were purchased from Boehringer Mannheim, BRG.
  • Porcine transforming growth factor ⁇ -type 1 (TGF- ⁇ 1) was obtained from R and D Systems, Minneapolis, Minn., USA.
  • the human histiocytic lymphoma cell line U937 (American Type Culture Collection (ATCC), CRL 1593) was obtained from Dr. A. Fattorssi (Research Lab. of Aeronautica Militare, Rome, Italy) and cultured in RPMI 1640 medium with 10% heat inactivated fetal calf serum and 2 mM L-glutamine at a density of 0.5 ⁇ 10 6 cells/ml at the onset of the experiment. The medium was supplemented with 100 units/ml of penicillin and 25 ug/ml streptomycin.
  • the human rhabdomyosarcoma (RD) and adenocarcinoma (A549) cell lines (ATCC CCL 136 and ATCC CCl 185, respectively) were obtained from Flow laboratories, Irvine, U.K., and kept in Dulbecco's modified Eagle's medium supplemented with 10% fetal calf serum until confluency, as described earlier (Lund et al., 1988). The cell lines were tested for and found free from Mycoplasma infection. PMA and the other compounds were present during different time periods and in varying concentrations, as indicated for each experiment. The adherent cells were released by a rubber policeman, and harvested for RNA analysis as described (Marcher et al., 1988).
  • the plasmid used as a probe for u-PAR mRNA (p-u-PAR-1) carries cDNA covering the entire coding region and the 3′- and the 5′-untranslated regions (Example 3).
  • the signal for u-PAR is extremely weak for the control cells but visible after longer exposure (result not shown). After 3 hours of PMA treatment, a visible signal for u-PAR mRNA is seen with a maximal effect after 24 hours of PMA treatment.
  • the Northern filter was stripped and rehybridized with a human ⁇ -actin cDNA probe (Ponte et al., 1983). No or only a little effect on the level of hybridization with the ⁇ -actin cDNA was seen after PMA treatment.
  • FIG. 18 shows a weak signal of 125 I-ATF cross-linked to the u-PAR in control U937 cells. After increasing time of PMA treatment both an increase in the strength of signal and a change to a lower electrophoretic mobility was seen.
  • FIG. 19 shows that u-PAR mRNA levels are increased after 48 hours of stimulation with PMA (150 nM), mEGF (20 ng/ml and TGF- ⁇ 1 7.5 ng/ml) in both A549 and RD cells.
  • PMA 150 nM
  • mEGF 20 ng/ml
  • TGF- ⁇ 1 7.5 ng/ml TGF- ⁇ 1 7.5 ng/ml
  • FIG. 20 shows a weak signal of 125 I-ATF cross-linked to the u-PAR in control U937 cells. After increasing time of dibuturyl cAMP treatment both an increase in the strength of signal and a change to a lower electrophoretic mobility was seen.
  • T7 and T3 polymerase pBluescriptKS(+) plasmid vector (Stratagene; CA, USA); RNasin and DNase I (Promega, WI, USA); [35]S-UTP (1300 Ci/mmol) (NEN Dupont, MA, USA); Dithiothretiol and restriction endonucleases (Boehringer Mannheim, Mannheim, FRG); K5 autoradiographic emulsion (Ilford, Cheshire, England); Formamide (Fluka, Buchs, Switzerland); Salmon Sperm DNA (Type III, Sigma, MO, USA). All other materials were as described previously (Kristensen et al., 1984; Kristensen et al., 1990), or of the best commercially available grade.
  • tissue preparation Following surgery, tissue specimens from 13 patients with adenocarcinoma of the colon were dissected and placed in 4% or 10% (wt/vol) formalin—0.9% NaCl solution for 24-48 hours before embedding in paraffin wax.
  • RNA probes Preparation of RNA probes. Fragments of the complete human u-PAR cDNA (see Example 3) were subcloned using standard techniques (Maniatis et al., 1982), and two subclones were prepared: pHUR04: PstI(184)-PstI(451) fragment and pHUR06: BamHI(497)-BamHI(1081) fragment in pBluescriptKS(+), base pair numbers corresponding to sequence as listed in Example 3. Pure plasmid preparations were prepared by banding in CsCl gradients and the plasmids were linearized for transcription using SmaI restriction endonuclease (pHUR04) or SpeI and EcoRI (pHUR06).
  • the pHUR04 template was transcribed with the T3 polymerase and the pHUR06 template linearized with EcoRI was transcribed with T7 polymerase, yielding antisense transcripts.
  • the pHUR06 template linearized by digestion with SpeI was transcribed with the T3 polymerase yielding sense transcripts.
  • In situ hybridization was performed using a method adapted from a number of published procedures (e.g. Cox et al., 1984; Angerer et al., 1987). Slides were dipped in 0.5% gelatin, 0.5% chrome-alum, dried at room temperature, baked at 180° C. for 3 hours and stored dust-free at room temperature. Paraffin sections were cut, placed on slides, heated to 60° C. for 30 minutes, deparaffinized in xylen and rehydrated through graded alcohols to PBS (0.01 M sodium phosphate buffer pH 7.4, containing 0.14 M NaCl). The slides were then washed twice in PBS, acid treated in 0.2 M HCl for 20 minutes and washed for 5 minutes in PBS.
  • PBS sodium phosphate buffer pH 7.4, containing 0.14 M NaCl
  • the probe was heated to 80° C. for 3 min and allowed to cool before addition to the hybridization mix.
  • the final hybridization solution contained RNA probe (80 pg/ ⁇ l), deionized formamide (50%), dextran sulphate (10%), t-RNA (1 ⁇ g/ ⁇ l), Ficoll 400 (0.02% (wt/vol), polyvinylpyrrolidone (0.02% (wt/vol)), BSA Fraction V (0.02% (wt/vol)), 10 mM DTT, 0.3 M NaCl, 0.5 mM EDTA, 10 mM Tris-Cl and 10 mM NaPO4 (pH 6.8).
  • the hybridization solution was applied to the slides (approx. 20 ⁇ l pr.
  • Sections were hybridized at 47° C. overnight (16-18 hours) in a chamber humidified with 10 ml of a mixture similar to the hybridization solution, except for probe, dextran sulphate, DTT and t-RNA (washing mix). After hybridization, the position of air bubbles occasionally formed over the section was marked, and coverslips were removed by incubation in washing mix for 1 hour at 50° C. The washing mix was changed, and washing continued for 1 hour at 50° C. Sections were washed in 0.5 M NaCl, 1 mM EDTA, 10 mM Tris-Cl (pH 7.2, NTE) with 10 mM DTT at 37° C.
  • Tissues were analyzed with antisense transcripts from the two non-overlapping clones pHUR04 and pHUR06 and with sense transcripts from pHUR06.
  • hybridization signal was consistently seen when using pHUR06 antisense transcripts.
  • a particularly prominent hybridization signal was found above cells at the leading edge of disrupted tumor glands in areas with clear signs of inflammation and degradation of surrounding mesenchymal tissue (FIGS. 21 A-B).
  • hybridization signal was located above cells closely associated with coherent strands of tumor cells (FIG. 21D) or above cells integrated at the serosal surface of the neoplastic epithelium itself (FIG. 21 C).
  • the hybridization signals obtained with pHUR06 antisense transcripts were confirmed on adjacent sections using antisense transcripts from pHUR04 (not shown). Unspecific binding of radioactive probe was demonstrated using sense transcripts from pHUR06 and in all tumors analyzed gave rise to a signal uniformly distributed above tissue sections and with an intensity comparable to that obtained with pHUR06 antisense transcripts in areas of no hybridization (e.g. normally appearing mucosa) (not shown).
  • Human fibrosarcoma cells (HT-1080, CCL 121) were obtained from the American Type Culture Collection, Rockville, Md. Confluent cell layers were grown in plastic Linbro wells (2 cm 2 ; Flow Laboratories) in Eagle's minimal essential medium (MEM) supplemented with 10% heat-inactivated (56° C. for 60 minutes) fetal calf serum (Gibco), 100 IU/ml penicillin and 50 ⁇ g/ml streptomycin. After reaching confluence, the cells were rinsed three times with MEM containing 0.2% bovine serum albumin (BSA), then changed to either serum-free medium (0.5 ml) or medium containing 10% heat-inactivated and plasminogen-depleted (i.e. absorbed with lysine-Sepharose; Pharmacia, Uppsala, Sweden) fetal calf serum as indicated in the Examples.
  • MEM Eagle's minimal essential medium
  • BSA bovine serum albumin
  • human plasmin (approximately 18 CU/mg; Kabi Diagnostica, Sweden) was added to the cultures at final concentrations of 0-5 ⁇ g/ml. The cells were incubated for 3 hours at 37° C. before assay of cell-bound and supernatant plasmin (see below). For plasmin release experiments, cells were loaded for 1 hour at 37° C. with 0-5 ⁇ g/ml plasmin in serum-free medium, then rinsed three times with MEM.
  • Human plasminogen (with glutamic acid N-terminal) was prepared by affinity chromatography on lysine-Sepharose (Deutsch, D. G., and E. T. Mertz, “Plasminogen: Purification from human plasma by affinity chromatography”, Science 170: 1095-1097, 1970) from freshly separated, unfrozen human plasma pretreated with 10 ⁇ M p-nitrophenyl guanidinobenzoate, 1 mM phenylmethylsulfonylfluoride and 0.1 ⁇ g/ml of an anti-catalytic murine monoclonal IgG antibody to human t-PA (ESP-2; see MacGregor, I. R.
  • ESP-2 anti-catalytic murine monoclonal IgG antibody to human t-PA
  • aprotinin Trasylol, Bayer, Leverkusen, FRG; 200 KIU/ml
  • tranexamic acid Cyclokapron, Kabi Vitrum, Swiss; 10 ⁇ M and 100 ⁇ M
  • human type-2 plasminogen activator inhibitor minactivin see Golder, J. P. et al., “Minactivin: A human monocyte product which specifically inactivates urokinase-type plasminogen activators”, Eur. J. Biochem. 136: 517-522, 1983
  • PAI-2 purified from cultures of human U-937 histiocytic lymphoma cells (see Leung, K-C.
  • DFP diisopropyl fluorophosphate
  • Active two-chain u-PA (Ukidan, Serono) was dissolved in 0.1 M Tris-HCl, pH 8.1, 0.1% Tween 80 (Tris/Tween).
  • Tris-HCl Tris-HCl
  • Tween 80 Tris/Tween 80
  • a freshly prepared solution of 500 mM DFP (Sigma) in isopropanol was added to yield a final DFP concentration of 5 mM. After thorough mixing, the sample was incubated for 2 hours at 37° C., after which period addition of DFP was repeated as above. After renewed incubation for 2 hours at 37° C., the reaction was terminated by thorough dialysis at 0° C. against Tris/Tween. No residual DFP inhibitor could be detected when the preparation was tested in an activity assay of soluble urokinase.
  • HT-1080 cells Confluent layers of HT-1080 cells were rinsed three times with methionine-free MEM medium containing 0.2% BSA, then prelabelled for 5 hours at 37° C. with 170 ⁇ Ci/ml ( 35 S)methionine (800 Ci/mmol, Amersham).
  • Human plasminogen 50 ⁇ g/ml
  • the neutralising monoclonal antibody to human PAI-1 10 ⁇ g/ml
  • Aprotinin 200 KIU/ml was added to both cultures before the medium was removed, after which the cells were rinsed three times with Dulbecco's medium containing 0.2% BSA.
  • the cell-bound u-PA was then eluted with 50 mM glycine/HCl (pH 3.0) containing 0.1 M NaCl for 3 minutes at 23° C. (Stoppelli et al., 1986).
  • the acid eluate was neutralised with 0.5 M Tris-HCl (pH 7.8) before immunoprecipitation for 2 hours at 23° C. with 3 ⁇ g/ml of goat IgG antibodies to human u-PA (American Diagnostica) or 3 ⁇ g/ml goat IgG antibodies to human t-PA (American Diagnostica) as control.
  • Immune complexes were collected by adsorption to protein A-Sepharose in an end-over mixer for 1 hour.
  • Immunoprecipitates were washed several times with immunoprecipitation buffer [10 mM Tris-HCl (pH 7.5), 50 mM NaCl, 0.5% sodium deoxycholate, 0.5% NP-40, 0.1% sodium dodecyl sulfate (SDS)] containing 100 KIU/ml aprotinin, twice with PBS and finally with 20 mM Tris-HCl (pH 7.5).
  • Immunocomplexes were solubilised by boiling in Laemmli's sample buffer (cf. Laemmli, supra) under reducing conditions (10% ⁇ -mercaptoethanol), and electrophoresed in 10% SDS-polyacrylamide gels. Fixed gels were treated with Amplify® (Amersham) and exposed to Kodak XAR-5 film at ⁇ 70° C.
  • Cell culture supernatants were assayed for pro-u-PA and active u-PA by the following modification of an immunocapture method (Stephens et al., 1988; Stephens et al., 1987).
  • Microtitre wells of polystyrene immunoplates (type 269620, A/S Nunc, Roskilde, Denmark) were coated overnight at 37° C. with 50 ⁇ l of a solution of goat IgG antibodies to human u-PA (cat. # 398, American Diagnostica).
  • the coating solution contained 2.5 ⁇ g of IgG per ml of 0.1 M sodium carbonate (pH 9.8).
  • the wells were treated with conditioned medium (50 ml) for 2 hours at 23° C., then rinsed again.
  • Half the wells were then treated with 50 ⁇ l of freshly prepared 2 ⁇ M p-nitrophenyl guanidinobenzoate (NPGB, Sigma) (Dan ⁇ , K., and E. Reich, “Plasminogen activator from cells transformed by an oncogenic virus—Inhibitors of the activator reaction”, Biochim. Biophys. Acta 566: 138-151, 1979) for 20 minutes at 37° C.
  • the other half (controls) received 50 ⁇ l of rinsing buffer (0.05% Tween 20 in PBS).
  • u-PA was assayed in all the wells by addition of 40 ⁇ l of plasminogen solution (100 ⁇ g/ml in assay buffer consisting of 50 mM sodium glycinate (pH 7.8), 0.1% Triton X-100, 0.1% gelatin and 10 mM 6-aminocaproic acid which also contained a very low concentration of plasmin (10 ng/ml)), and incubation took place for 30 minutes at 37° C. This concentration of plasmin in the plasminogen incubation was sufficient to enable full realization of the potential activity of pro-u-PA (cf. Petersen et al., 1988).
  • the plasmin produced by this incubation was assayed by its thioesterase activity (Green, G. D. G., and E. Shaw, “Thiobenzyl benzyloxycarbonyl-L-lysinate, substrate for a sensitive colorimetric assay for trypsin-like enzymes”, Anal. Biochem.
  • Pro-u-PA and active u-PA bound to the cell layer were recovered for immunocapture assays by the same method as was used in the metabolic labelling (see above). Each culture well (2 cm 2 ) was eluted with 150 ⁇ l of acid glycine at pH 3 (Stoppelli et al., 1986). For conditioned medium and cell-bound u-PA, the u-PA activity assayed after NPGB treatment was expressed as a percentage of the total activity obtained without NPGB treatment, and this percentage used as an index of pro-u-PA content (pro-u-PA index).
  • the plasmin activity of culture supernatant samples was assayed directly by incubation with the thioester substrate solution above (200 ⁇ l) for 30 minutes (serum-free supernatants) or 3 hours (serum-containing supernatants) at 37° C.
  • An estimate of the amount of active plasmin present was made from calibration curves using human plasmin dilutions in serum-free medium covering the appropriate ranges of activity.
  • Plasmin bound to the cell layer was recovered and assayed as follows. After harvest of culture medium, the cells were rinsed three times with PBS (plasmin assays of further rinses were negative); then the bound plasmin was specifically eluted (Miles, L. A., and E. F. Plow, “Binding and activation of plasminogen on the platelet surface”, J. Biol. Chem. 260: 4303-4311, 1985) with a solution of 1 mM tranexamic acid in the same rinsing solution (150 ⁇ l/well). Plasmin activity was assayed in eluate samples (50 ⁇ l) as above with an incubation time of 3 hours at 37° C. Tranexamic acid at 1 mM had no effect on the thioesterase activity of plasmin in these assays.
  • Plasminogen is Activated on the Cell Surface
  • plasmin activity could be recovered as a bound fraction from the cell layer.
  • the bound plasmin activity increased in a dose-dependent manner (FIG. 22 ).
  • the binding was specific so that after rinsing of the cells with isotonic buffer, the plasmin could be released by 1 mM tranexamic acid. This agent disrupts interactions with plasminogen or plasmin which involve the lysine affinity sites of the heavy-chain kringles (Miles, supra).
  • the plasmin released from HT-1080 cell surfaces was conveniently measured by its thioesterase activity, a method which was unaffected by the presence of tranexamic acid. Some plasmin activity was also detected in the medium.
  • a concentration of 40 ⁇ g/ml human plasminogen added to 0.5 ml of medium above a confluent 2 cm 2 cell layer activity corresponding to 28 ng of plasmin could be recovered from the cell layer with tranexamic acid, while 10 ng was measurable in the medium after 3 hours of incubation at 37° C. This concentration of plasminogen is well below the 200 ⁇ g/ml present in normal human plasma.
  • plasmin was added to the culture medium of HT-1080 cells. As shown in FIG. 23, virtually no plasmin activity was detected on the cell surface when the medium contained 10% fetal calf serum, while there was a considerable dose-dependent plasmin binding in the absence of serum.
  • HT-1080 cells are prolific producers of u-PA (Saksela, O., et al., “Plasminogen activators, activation inhibitors and alpha-2-macroglobulin produced by cultured normal and malignant human cells”, Int. J. Cancer 33: 609-616, 1984), but although they synthesize some t-PA, this does not appear to be secreted (R. Stephens, unpublished observations).
  • the activators were incubated with plasminogen in the presence of monoclonal antibodies that inhibit each of the activators.
  • u-PA is present both in the medium and bound to the cell surface (Nielsen et al., 1988).
  • the cells were preincubated with either the anti-catalytic u-PA antibody or PAI-2, and the cells were then thoroughly washed before being incubated with plasminogen in serum medium. Both inhibitors caused a significant decrease in the cell-bound plasmin activity, while no inhibition of the u-PA activity in the medium was detected (Table 6).
  • FIG. 25 An alternative method of studying the role of cell-bound versus free u-PA is illustrated in FIG. 25 .
  • u-PA is bound to its receptor, u-PAR, at the surface of HT-1080 cells (Nielsen et al., 1988). This binding does not involve the active site of u-PA (Blasi, F., 1988), and u-PAR therefore also binds u-PA that has been treated with the irreversible active-site titrant, DFP (Nielsen et al., 1988).
  • the HT-1080 cells were preincubated for 18 hours with DFP-inactivated u-PA which, when present in a large molar excess, resulted in a decrease of approximately 70% in surface-bound u-PA that was released by acid treatment (Stoppelli et al., 1986). Concomitantly, there was a comparable decrease in the amount of plasmin generated on the cell surface (FIG. 25 ).
  • u-PA is released into the medium of HT-1080 cells as a single-chain proenzyme, pro-u-PA, that can be converted to two-chain active u-PA by plasmin (Nielsen et al., 1982; Sim, supra).
  • the enzymatic activity of the proenzyme is at least 250-fold lower than that of the two-chain u-PA (Petersen et al., 1988), and it does not react with PAI-1 (Andreasen et al., 1986) or PAI-2 (Stephens et al., 1987; Wun et al., 1987).
  • pro-u-PA As an alternative way of distinguishing between pro-u-PA and active u-PA, the fact that low molecular weight active site reagents for u-PA do not bind to pro-u-PA (Nielsen et al., 1982) was utilized.
  • One of these, NPGB was used in a convenient test to distinguish between the two u-PA forms in an immunocapture assay (Stephens et al., 1988; Stephens et al., 1987).
  • the u-PA present in samples was first absorbed to u-PA antibodies bound to microtitre wells. Half the wells for each sample were treated with NPGB, the other half were control treated.
  • the total u-PA activity (pro-u-PA+active u-PA) in the untreated wells and the pro-u-PA in the treated wells were then measured by a coupled plasminogen activator assay in the presence of an initial concentration of 10 ng/ml plasmin, and the results were expressed as a pro-u-PA index.
  • FIG. 27A shows that the proportion of the total surface-bound u-PA that was present as pro-u-PA decreased dramatically when the cells were incubated with human plasminogen in serum medium in comparison with plasminogen-free cultures (from approximately 90% to approximately 10%).
  • the neutralizing PAI-1 antibody was therefore included in the next experiment in which the effect of the plasmin inhibitor aprotinin and the effect of an anti-catalytic monoclonal antibody to human plasmin on the conversion of pro-u-PA to active u-PA were studied. As shown in Table 7, both these inhibitors increased the relative amount of pro-u-PA, thus demonstrating that the activation of cell-bound pro-u-PA was catalyzed by plasmin.
  • Confluent cell layers in serum medium were preincubated for 1 hour at 37° C. with an anti-catalytic monoclonal antibody to human u-PA (10 ⁇ g/ml) or PAI-2 (titration equivalent of 3.6 IU u-PA/ml).
  • the cells were then rinsed three times with serum-free MEM medium before incubation for 3 hours at 37° C. with MEM medium containing 10% heat-inactivated and plasminogen-depleted fetal calf serum and native human plasminogen (Plg, 40 ⁇ g/ml).
  • the incubation with plasminogen was used as the 100% control for bound plasmin while the control for u-PA was the incubation without plasminogen.
  • Confluent cell layers were incubated for 2 hours at 37° C. with MEM medium (0.5 ml) containing 10% heat-inactivated and plasminogen-depleted fetal calf serum with the following additions: native human plasminogen (Plg, 40 ⁇ g/ml); neutralizing monoclonal antibody to human PAI-1 (10 ⁇ g/ml); aprotinin (200 KIU/ml); anti-catalytic monoclonal antibody to human plasmin (20 ⁇ g/ml); and tranexamic acid (TA, as shown).
  • Half the wells were then treated with aprotinin (200 KIU/ml) and used for assay of bound u-PA and its pro-u-PA index. The other half were used for elution and assay of bound plasmin.
  • PAI-1 was purified as described previously (Nielsen et al., 1986).
  • Pro-u-PA was purified from human A431 epidermoid carcinoma cells (Fabricant et al., Proc. Natl. Acad. Sci. USA 74, 565-569, 1977) as described by Corti et al. in Peptides of Biological Fluids (H. H. Peeters, Ed.), 33, 623-626, 1985, and was a kind gift from E. Sarubbi and A. Soffientini.
  • Human monocyte-like U937 cells derived from a histiocytic lymphoma (Sundstrom C. and Nilsson, K., Int. J. Cancer 17: 565-577, 1976) were grown in RPMI 1640 medium supplemented with 10% fetal calf serum.
  • u-PA/PAI-1 complex Preparation of u-PA/PAI-1 complex.
  • PAI-1 was activated before use by treatment with 4M guanidine-HCl for 1 hour at 37° C. (Hekman, C. M. and Loskutoff, D. J., J. Biol. Chem. 260, 11581-11587, 1985). Guanidine was removed by centrifugation through Centricon 10 (Amicon, Danvers, Mass.). The u-PA/PAI-1 complex was formed after incubation of the proteins at the ratios indicated in the results section for 1 hour at room temperature.
  • Iodinations 1 ⁇ g portions of protein (ATF, u-PA or pro-u-PA) in 30 mM sodium phosphate buffer (pH 7.4) were iodinated with 1 mCi of Na 125 I (Amersham Ltd., Amersham, UK) and 5 ⁇ g of Iodogen (Pierce Chemical Co., Rockford, Ill.) for 4 minutes at 4° C., and the reaction was stopped with excess N-acetyltyrosine. Iodinated proteins were separated from unincorporated radioactivity by gel filtration on Sephadex G-25. The specific activity obtained ranged within 80-150 ⁇ Ci/ ⁇ g protein. Iodinated u-PA was further purified by chromatography on benzamidine-Sepharose (Holmberg et al., 1976) to isolate molecules still retaining enzymatic activity.
  • Binding assay Before binding, U937 cells were incubated for 1 hour at 4° C. in RPMI 1640 medium supplemented with 0.1% bovine serum albumin and 50 mM Hepes (pH 7.4). The cells were then acid-treated in 50 mM glycine-HCl, 100 mM NaCl (pH 3) for 3 minutes at 4° C. and quickly neutralized with half a volume of 0.5 M Hepes, 100 mM NaCl (pH 7.4).
  • binding buffer phosphate buffered saline supplemented with 0.1% bovine serum albumin
  • iodinated ligands about 50,000 cpm corresponding to 0.1 nM for ATF and 0.05 nM for pro-u-PA and u-PA
  • binding buffer phosphate buffered saline supplemented with 0.1% bovine serum albumin
  • iodinated ligands about 50,000 cpm corresponding to 0.1 nM for ATF and 0.05 nM for pro-u-PA and u-PA
  • Plasmin cleavage of pro-u-PA 125 I-pro-u-PA was allowed to bind to cells as described above. After washing, the cells were incubated in the presence of plasmin (10 ⁇ g/ml) at room temperature for 10 minutes. Iodinated pro-u-PA in solution was activated under the same conditions. The reaction was stopped by the addition of aprotinin to a final concentration of 125 ⁇ g/ml.
  • u-PA activity was assayed by incubating 100 ⁇ l aliquots of binding mixtures or supernatants of binding assays in 0.05 M Tris-HCl (pH 7.5), 40 mM NaCl, 0.01% Tween 80, with 1 mM of the plasmin-specific substrate S2390 (Kabi Vitrum, Sweden) and 0.5 ⁇ M plasminogen in a final volume of 0.3 ml.
  • the time dependence of the colour development was measured following the absorbance at 405 nm (Petersen et al., 1988).
  • Plasminogen activator activity in electrophoretic gels was revealed by zymography (Granelli-Piperno, A. and E. Reich, J. Exp. Med. 148: 223-234 (1978)), layering the polyacrylamide gel over an agarose gel (1%) containing casein (2% non-fat dry milk) and plasminogen (40 ⁇ g/ml), in 255 mM Tris-HCl (pH 7.5).
  • the caseinolytic plaque assay was carried out essentially as described by Goldberg, A. R. (Cell 2: 95-102, 1974); briefly, U937 cells were resuspended in RPMI 1640 medium containing 0.8% agar, 1.3% non-fat dry milk, and 13 ⁇ g/ml plasminogen, and layered into a 30 mm plastic dish. After incubation for 3 hours at 37° C., the plates were visually scored and photographed. The whole plate was then dried and stained in 70% methanol, 10% acetic acid and 0.2% Coomassie blue.
  • PAI-1 PAI-1 with receptor-bound u-PA to assess if u-PA could, at the same time, interact with its inhibitor and its receptor, and if the two moieties of u-PA, the receptor-binding amino terminus and the inhibitor-binding, catalytically active carboxy terminus (Stoppelli et al., 1985), are completely independent.
  • the ability of PAI-1 to bind and inhibit receptor-bound u-PA would also demonstrate that the latter can be regulated mainly as the soluble u-PA and would strongly suggest that bound u-PA is catalytically active.
  • FIGS. 29A-B show the dependence of the inhibition of 125 I-ATF binding to U937 cells on the concentration of unlabelled u-PA or u-PA/PAI-1 complex.
  • preformed 125 I-u-PA/PAI-1 complexes were incubated with U937 cells for 1 hour at 4° C. After the binding step, the cells were wasehd and lysed, and the cell-associated radioactivity was analyzed by SDS-PAGE under non-reducing conditions. As shown in FIG. 30, in the absence of PAI-1 and of any unlabelled competitors, cell-bound radioactivity migrates mostly as a 50 kD band. However, with preformed u-PA/PAI-1 complexes, a cell-bound 90 kD band appears, corresponding to the migration of the u-PA/PAI-1 complex. This band represents receptor-bound u-PA/PAI-1 complex as it is competed for by unlabelled 85 nM ATF or u-PA.
  • FIG. 30 shows that 125 I-u-PA used for this experiment was in fact contaminated with 33 kD low molecular weight u-PA.
  • a 50 or 150 fold excess of PAI-1 much of the u-PA of the binding mixture is complexed to give 75 kD and 90 kD PAI-1 complexes, the former representing that with low molecular weight u-PA.
  • the 75 kD band is not found associated to U937 cells, which is in keeping with the notion that u-PA binds its receptor via the amino-terminal domain, which is missing in the low molecular weight u-PA.
  • u-PA binds the receptor through its amino-terminal extremity, and the binding is competed equally well by ATF or u-PA (Stoppelli et al., 1985). Accordingly, it was found that the binding of the u-PA/PAI-1 complex can be competed to the same extent by ATF and u-PA, with 50% competition reached around 1-2 nM (data not shown). Thus, even when complexed to its inhibitor, u-PA still binds specifically to its receptor.
  • pro-u-PA Plasmin activation of pro-u-PA coincides with the appearance of the 90 kD band, the intensity of which is proportional to the amount of added PAI-1. Also in this case, however, although weak, the cell-associated 90 kD band is observed in the absence of exogenous PAI-1. Since it only appears after pro-u-PA activation, it most likely represents a complex of u-PA with a plasminogen activator inhibitor.
  • u-PA/PAI-1 complex formation inhibits receptor-bound u-PA activity.
  • caseinolytic plaque assay was employed in which cells are plated in agar in the presence of plasminogen and casein. The presence of a plasminogen activator activity is visualized by the appearance of a clear plaque, due to the digestion of casein by plasmin.
  • FIGS. 32A-G caseinolytic plaques observed around individual U937 cells are shown, which are representative of the entire cell population. In all instances, plaque formation was plasminogen-dependent (not shown). Since U937 cells produce a very small amount of u-PA, some very small plaques are observed in the absence of exogenous enzyme (FIG.
  • U937 cells were first incubated with 10 nM DFP-u-PA for 60 minutes, washed and reincubated with active u-PA. Under these conditions, no activity can be detected (FIG. 32 E). Thus, the previously measured activity (FIG. 32B) can be competed by a u-PA analogue with a blocked active site. In the reverse experiment, first incubation with u-PA followed by a second incubation with DFP-u-PA, the latter does not prevent plaque formation (FIG. 32 G). Thus, very little dissociation of receptor-bound u-PA occurs during the time of the experiment.
  • PAI-1 is indeed capable of inhibiting the activity of receptor-bound u-PA, in suspension-growing U937 cells.
  • receptor-bound u-PA remains associated with the cell surface, is not detectably internalized or degraded, and can be dissociated from the surface by a mild acidic treatment (Stoppelli et al., 1985; Stoppelli et al., 1986; Vassalli et al., 1985). It was investigated whether receptor-bound u-PA/PAI-1 complex has a similar fate.
  • step 1 labelled ligands were incubated with acid-washed cells at 4° C. for 90 minutes (step 1). In all cases, more than 90% of the binding occurring during step 1 was inhibited by u-PA or ATF while no inhibition was obtained with low molecular weight u-PA, demonstrating the specificity of the interaction (data not shown).
  • step 2 the cells were incubated at 37° C. for 3 hours in binding buffer containing no ligands. The amount and the state of the ligand were assayed by quantitation of the radioactivity (and the extent of TCA precipitability) at different times during step 2. To this end, both the radioactivity recovered in the cell-associated form and in the supernatant were measured. The former was distinguished in radioactivity extracted by an acid wash (see Methods) representing receptor-bound ligand, and radioactivity resistant to the acid wash, representing cell-trapped internalized ligand (Haigler, 1980; Stoppelli et al., 1986).
  • u-PA two-chain u-PA
  • DFP-inactivated u-PA DFP-u-PA
  • ATF amino-terminal fragment of u-PA
  • PAI-1 preformed u-PA:PAI-1 complex.
  • the amount of receptor-bound ligand (cell-associated, acid-extracted radioactivity), of cell-trapped ligand (cell-associated, acid-resistant radioactivity) and of degraded (i.e. not precipitable by 10% TCA) ligand in the supernatant were measured at different times during step 2 incubation.
  • FIG. 33 shows the fate of the ligand during step 2 incubation at 37° C.
  • the receptor-bound fraction decreases slowly in agreement with previous data (Stoppelli et al., 1985); for u-PA, the decrease is somewhat faster.
  • the initial sharp loss of receptor-bound ligand continues throughout the incubation and after 2-3 hours at 37° C., very little complex is found still to be surface-bound.
  • the non-degraded, internalized ligand constitutes a small fraction in the case of ATF, but is clearly higher in all other cases.
  • the u-PA:PAI-1 complex it increases rapidly reaching about 40% of the total radioactivity around 30 minutes, and decreasing thereafter. While very little ligand is degraded in the case of ATF and DFP-u-PA, a larger fraction is degraded in the case of u-PA (20% after 3 hours) and much more in the case of the u-PA:PAI-1 complex (65% after 3 hours). In the latter case, the time course suggests a precursor-product relationship between the cell-trapped and the degraded ligand.
  • u-PA:PAI-1 complex but not ATF and DFP-u-PA, is internalized and then degraded.
  • u-PA might represent an intermediate case (low-level internalization and degradation) (see below).
  • the low-level degradation of u-PA might also reflect internalization of covalent u-PA:PAI-2 complexes since U937 cells produce this inhibitor.
  • the experiments were repeated including 50 nM low molecular weight u-PA during steps 1, 2 and in all washing buffers with the aim of titrating endogenous u-PA-binding PAI-like proteins. Quantitative data obtained under these conditions (FIG.
  • chloroquine is a drug inhibiting lysosomal protein degradation (De Duve et al., 1974; Carpenter and Cohen, 1976; McKanna et al., 1979).
  • a typical precursor-product relationship is observed in step 2 between the rate of accumulation of ligand in the pellet in a non-acid-extractable form and the rate of release of degraded ligand in the supernatant (in a TCA-soluble form). The latter reaches 60% of the total bound ligand after 3 hours.
  • free u-PA is apparently internalized and degraded by U937 cells, although at a slower rate (FIGS. 33 A-D). This is due to internalization of complexes formed between u-PA and endogenous proteins which interact with the u-PA active site, possibly the inhibitor PAI-2 (Vassalli et al., 1984; Genton et al., 1987).
  • the u-PA:PAI-1 complex is bound to the receptor itself (see Example 8) and subsequently undergoes internalization and degradation.
  • This receptor therefore, must alternate between two possible configurations: one in which it binds active u-PA and in which it dictates plasminogen activation on the cell surface; and another in which it binds the inhibited enzyme and in which it favours internalization and degradation of the ligand. This property could be exploited for internalizing toxins and thus specifically kill the cells that express the u-PA receptor, or by forcing the state of the receptor from one state (i.e. exposed) to another, through PAI-1 or PAI-1 analogues.
  • Plasminogen was purified from fresh human plasma as previously described (Dan ⁇ and Reich, 1979), and was further separated into its two isoforms by elution from lysine-Sepharose with a linear gradient of 6-amino-hexanoic acid. Plasminogen isoform 2 was used in all experiments described here.
  • u-PA M r 55,000
  • pro-uPA Ellis et al., 1987
  • Ukidan Serono
  • the concentration of active u-PA in these preparations was determined by active-site titration with p-nitrophenyl-p-guanidinobenzoate (Sigma Chem. Co.).
  • DFP-inactivated u-PA was prepared as described in Example 1.
  • the murine monoclonal antibody to u-PA was clone 2 from Nielsen et al., 1986.
  • Active PAI-1 was purified from the serum-free conditioned medium of Hep G2 cells by affinity chromatography on immobilized anhydro-urokinase (Wun et al., 1989).
  • PAI-2 was purified from U937 cell lysates by chromatofocusing as described (Kruithof et al., 1986).
  • the concentrations of active inhibitor in the various PAI preparations were determined by titration against u-PA immediately before use in the kinetic experiments.
  • PAI-1 or PAI-2 at varying concentrations between 1 nM and 100 nM were incubated with active-site titrated u-PA (20 ⁇ M) for 1 hour at 37° C. in 0.05 M Tris, 0.1 M NaCl pH 7.4 containing 0.2% bovine serum albumin. Residual u-PA activity was then measured by hydrolysis of 0.2 mM Glu-Gly-Arg-AMC (Bachem, Switzerland).
  • U937 cells were grown in suspension in RPMI 1640 medium supplemented with 5% heat-inactivated fetal calf serum.
  • PMA-stimulation of U937 cells was performed at a cell density of 0.5 ⁇ 10 6 cells/ml with 150 nM PMA for 4 days.
  • the adherent cell population was harvested with a rubber scraper and resuspended in PBS.
  • the cells were then incubated at a final concentration of 1 ⁇ 10 6 cells/ml in 0.05 M Tris-HCl, 0.1 M NaCl with plasminogen 2 (0.175 ⁇ M) and 0.2 mM of the plasmin specific fluorogenic peptide substrate H-D-Val-Leu-Lys-AMC (Bachem, Switzerland). These incubations were made in 10-mm plastic fluorimeter cuvettes which were maintained at 37° C. and gently stirred in a Perkin-Elmer LS-5 spectrofluorimeter equipped with a micro magnetic stirrer.
  • the fluorescence was measured at 1 minute intervals at an excitation wavelength of 380 nm and an emission wavelength of 480 nm, with both slits set to 5 nm. These data were converted to plasmin concentrations by calculating the rate of change in fluorescence between each time point and comparison with a calibration curve constructed using active-site titrated plasmin.
  • PAI's The effect of PAI's on the activity of cell-bound u-PA was determined by the addition of varying concentrations of PAI-1 (0.18-18.4 nM) or PAI-2 (1.13-56.7 nM) to the incubations at the same time as the addition of plasminogen. Curves were then constructed of plasmin concentration against time.
  • k app is the apparent pseudo-first order rate constant and [pln] t and [pln] ⁇ , respectively, are the plasmin concentrations at time, t and at infinite time when u-PA is completely inhibited.
  • ⁇ [pln] 0 is the initial rate of plasmin generation which was determined in control incubations in the absence of inhibitors.
  • the experimentally obtained plasmin generation curves were fitted to equation 1 by non-linear regression.
  • the identity of the bound enzyme was confirmed as u-PA, rather than tPA or unidentified activator, by its complete inhibition by an anticatalytic monoclonal antibody to u-PA (FIG. 36 ).
  • Incubation of the cells with exogenously added u-PA resulted in an increased rate of plasminogen activation (FIG. 36 ), due to saturation of the previously unoccupied receptors.
  • the endogenously-bound u-PA could be eluted from the cells by brief acid treatment (Stoppelli et al., 1986) and the cells then saturated with exogenously added u-PA, which gave rise to an approximately 50% higher rate of plasmin generation (FIG. 36 ).
  • the binding of u-PA to the cells could be competed by preincubation of the acid-washed cells with a 100-fold molar excess of DFP-inactivated u-PA (87% inhibition of plasminogen activation), demonstrating that the binding was via specific interaction with the u-PA receptor. This was further demonstrated by preincubation of the cells with 25 ug/ml of a polyclonal antibody raised against the purified human u-PA receptor, which resulted in 82% inhibition of plasminogen activation as detailed in Example 10. Pre-immune IgG from the same animal in which this antibody was raised had no effect on u-PA binding. In control experiments cells prepared by each of the above methods were found to be indistinguishable with respect to their inhibition by PAI's, therefore the studies subsequently described were performed using cells with endogenously-bound u-PA.
  • PAI-1 used for these studies was purified from the conditioned medium of Hep G2 cells, and in contrast to PAI-1 preparations purified from other cell-types does not require pre-treatment with denaturants for inhibitory activity.
  • This preparation contains the NH 2 -terminal fragment(s) of vitronectin which may be reponsible for the stabilization of the PAI-1 activity.
  • the effect of this PAI-1 preparation on plasminogen activation by cell-bound u-PA is shown in FIG. 37 .
  • PAI-1 inhibits u-PA catalyzed plasminogen activation on the cell-surface in a time and concentration dependent manner and also that at the higher concentrations of PAI-1 used there is complete inhibition of u-PA activity within the time-course of the experiments.
  • These data give an association rate constant for the inhibition of cell-bound u-PA by PAI-1 of 4.5 ⁇ 10 6 M ⁇ 1 s ⁇ 1 (FIG. 39 ).
  • the inhibition by PAI-1 of plasminogen activation by u-PA in solution was determined as 7.9 ⁇ 10 6 M ⁇ 1 s ⁇ 1 (FIG.
  • Vitronectin is able to interact with cells through an Arg-Gly-Asp adhesion sequence and thereby promote cell attachment and spreading. This sequence is apparently still available in PAI-1/vitronectin complexes (Salonen et al., 1989) and may therefore be playing a role in the inhibition of cell-bound u-PA.
  • U937 cells were pre-incubated with the peptide Gly-Arg-Gly-Asp-Ser (SEQ ID NO: 27) (0.5 mg/ml) for 30 minutes prior to incubation with the PAI-1 preparation (0.9 or 4.5 nM). The rates of inhibition of cell-bound u-PA were found to be the same in the presence or absence of the peptide.
  • FIG. 38 shows the inhibition of cell-bound u-PA by varying concentrations of PAI-2. There is once again a concentration and time dependent inhibition of plasminogen activation, and complete inhibition of plasminogen activation was observed within the time-course of the experiment at the higher concentrations of PAI-2.
  • PAI-2 inhibits cell-bound u-PA with an association-rate constant that is approximately 60% of that obtained for u-PA in solution, which is very similar to the effect observed with PAI-1. Therefore, similarly to PAI-1, PAI-2 is virtually as efficient an inhibitor of cell-bound u-PA as it is of u-PA in solution.
  • PAI-1 was found to inhibit cell-bound u-PA with a lower association-rate constant than on unstimulated cells, 1.7 ⁇ 10 6 M ⁇ 1 s ⁇ 1 compared to 4.5 ⁇ 10 6 M ⁇ 1 s ⁇ 1 (Table 9), which represents approximately 20% of the rate of inhibition of u-PA in solution.
  • the inhibition of cell-bound u-PA by PAI-2 was also reduced to a similar extent upon PMA stimulation of the cells, from 3.3 ⁇ 10 5 M ⁇ 1 s ⁇ 1 to 1.1 ⁇ 10 5 M ⁇ 1 s ⁇ 1 (Table 9).
  • Plasminogen activation by u-PAR-bound u-PA on U937 cells was determined as described in detail in Example 9. Briefly, varying concentrations of plasminogen (0.09 ⁇ M and 2.26 ⁇ M) were incubated with U937 cells (pre-incubated with active u-PA and subsequently washed) in the presence of the plasmin specific fluorogenic substrate Val-Leu-Lys-AMC (Bachem, Switzerland). Plasmin generation was determined from the rate of change of the increase in fluorescence due to substrate hydrolysis, measured at excitation and emission wavelengths of 380 nm and 480 nm, respectively.
  • the concentration of u-PA bound to u-PAR on U937 cells was determined using 125 I-u-PA (prepared as described in Example 8) which was incubated with the cells in parallel incubations to the kinetic experiments and treated identically. 125 I-u-PA bound to u-PAR was then quantitated using standard gamma-counting techniques.
  • u-PA activity was determined by the addition of an equal volume of a solution containing 0.2 mg/ml Glu-plasminogen (Kabi, Sweden) and 0.2 mM Val-Leu-Lys-AMC in 0.05 M Tris pH 7.4, 0.1 M NaCl, 2 mM trans-4-(aminomethyl)-cyclohexane-carboxylic acid (Sigma Chem. Co.). Hydrolysis of the fluorogenic substrate was monitored continuously in a Perkin-Elmer LS5 spectrofluorimeter with the excitation and emission wavelengths set at 380 nm and 480 nm, respectively. Residual u-PA concentrations were calculated from these data by reference to standard curves constructed using u-PA of known concentration.
  • u-PA bound to u-PAR on U937 cells was found to activate its natural substrate plasminogen with different kinetic characteristics from those displayed in the absence of u-PAR.
  • the activation of Glu-plasminogen by u-PAR-bound u-PA followed an apparently Michaelis-Menton type kinetic mechanism. This was characterized by a K m of 0.67 ⁇ M and a k cat of 5.6 min ⁇ 1 (FIG. 40 ). Both of these constants were different from those obtained with u-PA in solution, i.e. the absence of U937 cells.
  • plasminogen As plasminogen (and plasmin) is known to bind to U937 cells, as well as a wide variety of other cells (Ellis et al., 1989; Plow et al., 1986), these constants measure plasminogen activation taking place at the surface of cells possessing u-PAR, i.e. cell-surface plasminogen activation.
  • Table 10 also shows similar data for plasminogen activation by u-PA bound to u-PAR on PMA-stimulated U937 cells.
  • the K m for plasminogen activation is now 1.43 ⁇ M, still much lower than for the reaction in solution.
  • the k cat also falls from 5.6 min ⁇ 1 to 1.23 min ⁇ 1 , resulting in an overall reduction in plasminogen activation (k cat /K m ) of approximately 10-fold when compared to unstimulated cells.
  • the polyclonal rabbit antibody raised against purified u-PAR was used to demonstrate that the cell-surface plasminogen activating activity of u-PA demonstrated in the previous section was indeed due to u-PA binding to u-PAR, and also to demonstrate that this antibody did block binding of u-PA to u-PAR in solution.
  • the anti-u-PAR antibody When pre-incubated with U937 cells at a concentration of 25 ⁇ g/ml for 30 minutes, the anti-u-PAR antibody resulted in a decrease in the subsequent plasminogen activating activity of 76% (mean of three experiments, range 66%-82%). In contrast the preimmune IgG gave ⁇ 1% inhibition, whilst DFP-u-PA gave 90% inhibition (range 74%-100% in three experiments). Therefore the anti-u-PAR polyclonal antibody effectively inhibits the cell-surface plasminogen activation.
  • PMA-stimulated U937 cells were treated with PI-PLC for 120 minutes.
  • Supernatants from both treated and control cells were incubated with monoclonal antibodies to PAI-2.
  • 20 ⁇ l of supernatant was incubated with u-PA in a final volume of 100 ⁇ l.
  • mice of the BALB/c strain were immunized with u-PAR purified on a diisopropylfluoride urokinase-type plasminogen activator (DFP-u-PA) ligand affinity column.
  • DFP-u-PA diisopropylfluoride urokinase-type plasminogen activator
  • Myeloma cells and X63-Ag 8.653 cells (Kearney, J. Immunol. 123: 1548-1550, 1979) in logarithmic phase of growth were isolated for fusion with BALB/c spleen lymphocytes. The myeloma cells were resuspended in serum-free medium.
  • spleen lymphocytes and myeloma cells were mixed in a ratio of 1:1.
  • HAT hypoxanthine-aminopterin-thymidine
  • the selection medium was renewed after a week or when needed.
  • HAT medium 10-14 days after fusion, HAT medium was replaced by HT medium and later, e.g. after 10 days, by regular medium.
  • Hybrid cells secreting the desired antibodies were frozen in liquid N 2 as early as possible.
  • Radioimmunoprecipitation assay (RIPA). This assay as well as the reverse solid phase radioimmune assay were developed because the amount of purified antigen was limited.
  • Reaction buffer 0.1% bovine serum albumin+0.1% Triton X-100 in PBS (0.1% BSA, 0.1% Triton X-100/PBS).
  • Coating buffer 0.1 M Na 2 CO 3 , pH 9.8.
  • Blocking buffer 25% fetal calf serum in PBS (25% FCS/PBS).
  • Washing buffer PBS+0.1% Tween 20, pH 7.4 (PBS/Tween 20).
  • HRP-REM Ig Horseradish peroxidase-conjugated rabbit anti-mouse Ig
  • Blocking buffer 25% fetal calf serum in PBS (25% FCS/PBS) or 1% skimmed milk powder (SMP) in PBS.
  • Citrate buffer 0.1 M citrate, pH 5.0.
  • Substrate solution 1,2-Phenylenediamine dihydrochloride (OPD) tablets in citrate buffer, e.g. 3 OPD tab. in 15 ml of citrate buffer+5 ⁇ l of H 2 O 2 (30%).
  • OPD 1,2-Phenylenediamine dihydrochloride
  • Stop buffer 1 M H 2 SO 4 .
  • Samples of purified human u-PA receptor (Example 1) were subjected to SDS-polyacrylamide gel electrophoresis under non-reducing conditions on a 6-16% gradient gel. By the use of fluorescent molecular weight markers run in neighbouring lanes, the electrophoretic region corresponding to the antigen was excised. The gel piece was lyophilized and subsequently macerated in a Mikro-Dismembrator II apparatus (B. Braun AG, Federal Republic of Germany). The polyacrylamide powder was reconstituted in Tris-buffered saline, mixed with Freund's incomplete adjuvant and used for injection of a New Zealand white rabbit.
  • the animal received 5 injections, each containing approximately 3 ⁇ g of the antigen, over a 10 week period, followed by a single 8 ⁇ g injection after an additional 7 weeks. Serum was drawn 1 week after the last injection, and IgG was prepared by Protein A-Sepharose chromatography. In order to remove antibodies against trace impurities in the injected antigen, the antibody was absorbed by consecutive passages through columns containing immobilized human u-PA and the protein mixture constituting the Triton X-114 detergent phase from PMA-stimulated U937 cells (see Example 1), respectively. The antibody preparation obtained did not inhibit the amidolytic or plasminogen activator activity of u-PA in solution.
  • Electrophoresis SDS-PAGE was carried out in slab gels with a linear 6-16% polyacrylamide concentration gradient according to Laemmli (supra). Samples were run under reducing conditions. The samples were reduced immediately before electrophoresis in Laemmli buffer except that 2-mercaptoethanol was replaced with dithiothreitol for 3 minutes at 100° C.
  • molecular weight markers were used: phosphorylase b (molecular weight about 94,000), bovine serum albumin (molecular weight about 67,000), ovalbumin (molecular weight about 43,000), carbonic anhydrase (molecular weight about 30,000), soybean trypsin inhibitor (molecular weight about 20,100), and ⁇ -lactalbumin (molecular weight about 14,400).
  • the sheets were rinsed, incubated with secondary antibody (alkaline phosphatase-conjugated rabbit anti-mouse Ig (Dakopatts, Copenhagen)), and developed with nitro blue tetrazolium/5-bromo-4-chloro-3-indolyl phosphate/Levamisol.
  • secondary antibody alkaline phosphatase-conjugated rabbit anti-mouse Ig (Dakopatts, Copenhagen)
  • Western blotting analysis of rabbit u-PAR antibody was performed in the same manner, except for the following modifications: SDS-PAGE was performed under non-reducing conditions. Newborn calf serum was used instead of fetal calf serum. Only 10% serum was included in the primary antibody incubation step. Alkaline phosphatase conjugated swine anti-rabbit Ig (Dakopatts code 306), 100-fold dilution, was used as the secondary antibody.
  • the final concentration of 125 I-ATF was 2.2 nM, and the final dilutions of anti-u-PAR serum/control serum ranged from 1:300 to 1:153,600.
  • the cells were then washed 3 times with 1 ml of PBS-bovine serum albumin, and the bound radioactivity was measured in a gamma counter. Under these conditions, 12% of the radioactivity became cell-bound when no antiserum had been added. 90% of the bound radioactivity was displaced when the cells were preincubated with 700 nM non-labelled u-PA.
  • the antiserum was used to immunocapture 125 I-labelled purified u-PAR (FIG. 43 ).
  • a 2-fold serial dilution of the anti-u-PAR serum 1:500-1:32000 showed that the same amount of 125 I-u-PAR (about 2% of total) was captured at a serum dilution up to 1:4000 and dropped to half the amount at 1:32000.
  • the same serial dilution of non-immune serum and the other controls resulted in a capture of 125 I-u-PAR of about 0.5% of total.
  • the reaction of immune versus non-immune serum in an ELISA is shown in FIG. 44. 1 ng of purified u-PAR coated per well was sufficient to be detected with the immune serum diluted 1:8000. Both the non-immune serum at all dilutions and other controls gave reaction values at background level.
  • the mouse antiserum against human u-PAR was used in a competition experiment in which U937 cells were preincubated with the antiserum followed by addition of 125 I-ATF. As shown in FIGS. 45A-B, the anti-u-PAR serum was able to completely inhibit the specific binding of 125 I-ATF to the cells. 50% inhibition was obtained at a 1:2400 dilution. Under the same conditions, a control serum showed only slight inhibition, i.e. about 20% at the highest concentration used (a 1:300 dilution). In Western blotting, the u-PAR contained within the detergent phase from PMA-treated U937 cells, as well as the purified u-PAR, were detected by the anti-u-PAR serum (FIG. 45B, lanes 1 and 2). The control immune serum gave no reaction with the same preparations (lanes 3 and 4).
  • Rabbit polyclonal antibodies were prepared by immunizing a rabbit with polyacrylamide gel material containing affinity-purified u-PAR that had subsequently been subjected to preparative SDS-PAGE.
  • the IgG fraction was isolated from the obtained antiserum and absorbed by passage through columns with immobilized human u-PA and immobilized membrane-protein mixture derived from PMA-stimulated U937 cells, respectively.
  • the antibody recognized u-PAR in the Triton X-114 detergent phase from PMA-stimulated U937 cells (FIG. 46 A).
  • the effect of the rabbit antibody on the ligand binding capability of u-PAR was studied in a different experiment (not shown) in which a purified sample of u-PAR (Example 1; approximately 20 ng/ml) was preincubated with the purified and absorbed IgG from the rabbit anti-u-PAR serum (final IgG concentration 90 ⁇ g/ml during preincubation). This treatment completely hindered the subsequent formation of cross-linked conjugates with 125 I-ATF. The IgG from the pre-immune serum had no effect on the cross-linking assay at the same concentration.
  • u-PA was purified by affinity chromatography on monoclonal antibodies and activated by treatment with plasmin (Nielsen et al., 1982). Alternatively, two-chain u-PA was obtained commercially (Serono u-PA). u-PA was DFP-inactivated as described for the preparation of columns for purification of the receptor.
  • u-PA was dialyzed overnight against 0.1 M Na 2 HCO 3 with 0.1% Triton X-100.
  • N-biotin-hydroxysuccinimide was dissolved in N,N-dimethylformamide (5 mM).
  • To the u-PA preparation was added 0.1 ⁇ l of this solution per ⁇ g of u-PA, and the reaction was allowed to run for 1 hour at room temperature. Excess labelling compound was removed by dialysis overnight against 0.1 M NaHPO 4 , pH 8.0, with 0.5 M NaCl and 0.1% Triton X-100.
  • Cultured cells (PMA-treated U937) or cryostat sections of freshly frozen human chorion were treated for 3 minutes at room temperature with 0.05 M glycine, pH 3.0 with 0.1 M NaCl, neutralized with 0.5 M HEPES, pH 7.5 with 0.1 M NaCl and incubated at 4° C. with 200 nM of biotinylated DFP-treated u-PA dissolved in PBS with 0.1% BSA (PBS-BSA). Competition experiments were performed by simultaneous incubation with biotinylated DFP-treated u-PA (200 nM) and purified unlabelled u-PA (2 ⁇ M).
  • the u-PA receptor could be visualized on the surface of U937 cells, as shown in FIG. 47 A.
  • the signal was particularly prominent over cellular extensions. Incubation with a surplus of unlabelled u-PA showed competition of the signal (FIG. 47 B).
  • u-PA Human 54 kDa two-chain u-PA (Ukidan, Serono) was preincubated with u-PAR or buffer at the concentrations indicated for 15 min at room temperature. Plasminogen (10 ⁇ g/ml final concentration) and H-D-Valyl-L-leucyl-L-lysine-p-nitroanilide dihydrochloride (Kabi product S-2251), termed substrate S1 below (final concentration 400 ⁇ M) were added in a final reaction volume of 250 ⁇ l, and cleavage of the substrate was followed during incubation at 37° C. Standard curves were drawn from assays of the following final concentrations of u-PA: 8, 16, 32, 64, 128 and 256 pg/ml.
  • Human pro-u-PA was preincubated with u-PAR or buffer for 10 min at room temperature. Plasmin (10 ng/ml final concentration) was added and the samples were incubated at 37° C. Aliquots were taken after the following periods of incubation: 1, 2, 5, 10, 20, 30 and 60 min. After the periods indicated, plasmin activity within each sample was stopped by the addition of Trasylol (10 ⁇ g/ml final concentration).
  • the u-PAR concentration used led to an apparent 50% inhibition of u-PA activity in the dynamic u-PA concentration range of 32-256 pg/ml.
  • the standard curves obtained in the presence and absence of u-PAR were superimposable according to a model in which the activity of any u-PA concentration in the presence of u-PAR was equivalent to the activity of 50% of the same concentration in the absence of u-PAR.
  • the generated u-PA activity in each aliquot was measured spectrophotometrically after addition of the chromogenic u-PA substrate (S2) (see Methods; 400 ⁇ M final concentration) and incubation for 19 h at 37° C.
  • the activity was expressed as the equivalent concentration of commercial two-chain u-PA (see Methods), as read from a standard curve drawn from a simulteneous and parallel experiment.
  • the curves of u-PA activity vs. time of plasmin treatment were linear in the range from 2-20 min.
  • u-PA activity was generated at a velocity of 0.60 ng/ml equivalent two-chain u-PA per min.
  • the activation velocity was reduced to 0.18 ng/ml equivalent two-chain u-PA per min. This reduction was due to a real inhibition of pro-u-PA activation since the presence of u-PAR had no effect on the activity of two-chain u-PA against the substrate S2.
  • Urokinase-type plasminogen activator proenzyme, receptor, and inhibitors. J Cell Biol 104: 801-804
  • Urokinase-type plasminogen activator (u-PA) antigen is a predictor of early relapse in breast cancer. Fibrinolysis 1-10
  • Transforming growth factor- ⁇ is a strong and fast acting positive regulator of the level of type-1 plasminogen activator inhitor mRNA in WI-38 human lung fibroblasts. EMBO J 6: 1281-1286
  • the major Fc receptor in blood has a phosphatidylinositol anchor and is deficient in paroxysmal nocturnal haemoglobinuria. Nature 333: 565-567

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Public Health (AREA)
  • Pathology (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Biotechnology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Hospice & Palliative Care (AREA)
  • General Physics & Mathematics (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Toxicology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
US08/442,108 1989-04-07 1995-05-16 Urokinase-type plasminogen activator receptor Expired - Lifetime US6248712B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US08/442,108 US6248712B1 (en) 1989-04-07 1995-05-16 Urokinase-type plasminogen activator receptor

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US33461389A 1989-04-07 1989-04-07
US37485489A 1989-07-03 1989-07-03
US08/319,052 US5891664A (en) 1989-04-07 1994-10-06 Vectors and methods for recombinant production of uPA-binding fragments of the human urokinase-type plasminogen receptor (uPAR)
US08/442,108 US6248712B1 (en) 1989-04-07 1995-05-16 Urokinase-type plasminogen activator receptor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/319,052 Division US5891664A (en) 1989-04-07 1994-10-06 Vectors and methods for recombinant production of uPA-binding fragments of the human urokinase-type plasminogen receptor (uPAR)

Publications (1)

Publication Number Publication Date
US6248712B1 true US6248712B1 (en) 2001-06-19

Family

ID=26989284

Family Applications (1)

Application Number Title Priority Date Filing Date
US08/442,108 Expired - Lifetime US6248712B1 (en) 1989-04-07 1995-05-16 Urokinase-type plasminogen activator receptor

Country Status (9)

Country Link
US (1) US6248712B1 (de)
EP (2) EP0467932B1 (de)
JP (2) JPH04506148A (de)
AT (1) ATE197067T1 (de)
AU (2) AU652917B2 (de)
CA (1) CA2051676A1 (de)
DE (2) DE69033653T2 (de)
DK (1) DK0467932T3 (de)
WO (1) WO1990012091A1 (de)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060008535A1 (en) * 2004-07-09 2006-01-12 Robert Sabin Anti tumor compositions and methods of use
WO2005048822A3 (en) * 2003-11-18 2006-12-07 Attenuon Llc Antibodies and/or conjugates thereof which bind to the amino terminal fragment of urokinase, compositions and uses thereof
US20080081051A1 (en) * 2006-09-28 2008-04-03 Robert Sabin Method of manufacturing anti-tumor and anti-viral compositions
WO2008077958A2 (en) * 2006-12-22 2008-07-03 Hvidovre Hospital Soluble urokinase plasminogen activator receptor (supar) as marker for diseases
US20080199476A1 (en) * 2006-12-08 2008-08-21 Graham Parry Urokinase-type plasminogen activator receptor epitope, monoclonal antibodies derived therefrom and methods of use thereof
WO2008115870A2 (en) * 2007-03-16 2008-09-25 Mount Sinai School Of Medicine Induction and/or maintenance of tumor dormancy by disruption of urokinase plasminogen activator receptor-integrin interaction
US20080280833A1 (en) * 2005-01-31 2008-11-13 Bernard Degryse Therapeutic Peptides Derived from Urokinase Plasminogen Activator Receptor
US20090004176A1 (en) * 2001-11-02 2009-01-01 Paion Deutschland Gmbh Non-neurotoxic plasminogen activating factors for treating of stroke
EP2126067A2 (de) * 2007-02-23 2009-12-02 Landing Biotech, Inc. Atherosklerose-behandlung
US20100075372A1 (en) * 2006-09-28 2010-03-25 Taka-Aki Sato Method for deparaffinization of paraffin-embedded specimen and method for analysis of paraffin-embedded specimen
EP2390308A1 (de) 2002-02-25 2011-11-30 Vaxiion Therapeutics Inc Minizell-Zusammensetzungen und Verfahren
WO2015048619A3 (en) * 2013-09-30 2015-05-28 Teleflex Medical Incorporated Stabilized enzyme compositions
US9333280B2 (en) 2009-02-25 2016-05-10 Teleflex Medical Incorporated Stabilized enzyme compositions
WO2020140101A1 (en) 2018-12-28 2020-07-02 Catalyst Biosciences, Inc. Modified urokinase-type plasminogen activator polypeptides and methods of use
US11613744B2 (en) 2018-12-28 2023-03-28 Vertex Pharmaceuticals Incorporated Modified urokinase-type plasminogen activator polypeptides and methods of use

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2246779B (en) * 1990-08-03 1994-08-17 Delta Biotechnology Ltd Tumour-associated protease inhibitors targeted to tumour cells
AU661978B2 (en) * 1990-10-18 1995-08-17 Cancerforskningsfondet Af 1989 Antibodies against the urokinase receptor and their use
GB9026737D0 (en) * 1990-12-08 1991-01-30 Delta Biotechnology Ltd Cancer treatment
US5679350A (en) * 1992-05-28 1997-10-21 The University Of Toledo Method of delivery of a medicament to a cancer cell using a pathway of plasminogen activator material
ES2143504T3 (es) * 1992-05-28 2000-05-16 Univ Toledo Uso de un material citotoxico acoplado a un material inhibidor del activador plasminogeno para la produccion de un medicamento para el tratamiento de celulas cancerigenas.
JP2852192B2 (ja) * 1994-07-08 1999-01-27 カンセアフォースクニングスフォンデン・アフ・1989(フォンデン・チル・フレメ・アフ・エクスペリメンテル・カンセアフォースクニング) uPARのドメイン2+3のuPA結合部位および抗体
IT1271684B (it) * 1994-07-22 1997-06-04 Consiglio Nazionale Ricerche Oligonucleotidi antisenso complementari all'rna messaggero del recettore umano della urochinasi
US6277818B1 (en) 1998-10-29 2001-08-21 Angstrom Pharmaceuticals, Inc. Cyclic peptide ligands that target urokinase plasminogen activator receptor
US5942492A (en) * 1996-11-12 1999-08-24 Angstrom Pharmaceuticals, Inc. Cyclic peptides that bind to urokinase-type plasminogen activator receptor
US5994309A (en) 1997-07-25 1999-11-30 Angstrom Pharmaceuticals, Inc. Anti-invasive and anti-angiogenic compositions and methods
US6896870B1 (en) 1999-10-01 2005-05-24 Angstrom Pharmaceuticals, Inc. Diagnostic probes and therapeutics targeting uPA and uPAR
WO2001025410A2 (en) * 1999-10-01 2001-04-12 Angstrom Pharmaceuticals, Inc. DIAGNOSTIC PROBES AND THERAPEUTICS TARGETING uPA AND uPAR
US6649597B1 (en) 2000-06-22 2003-11-18 The University Of Iowa Research Foundation Targeting vector to the urokinase plasminogen activator receptor
US7045283B2 (en) * 2000-10-18 2006-05-16 The Regents Of The University Of California Methods of high-throughput screening for internalizing antibodies
AU2002229721A1 (en) * 2001-01-25 2002-08-06 Fondazione Centro San Raffaele Del Monte Tabor Methods and agents modulating upa/upar activity
KR100871454B1 (ko) 2001-05-21 2008-12-03 옴릭스 바이오파머슈티컬즈 에스. 에이. 단백질 용액으로부터 플라스민(오겐)의 제거
JP2004137151A (ja) * 2002-10-15 2004-05-13 Keio Gijuku 脳腫瘍の治療・診断薬
TW200813091A (en) * 2006-04-10 2008-03-16 Amgen Fremont Inc Targeted binding agents directed to uPAR and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4571421A (en) * 1979-11-05 1986-02-18 Genentech, Inc. Mammalian gene for microbial expression
WO1986006100A1 (en) * 1985-04-15 1986-10-23 Biotechnology Research Partners, Limited Human anti-inflammatory phospholipase inhibitor protein
US4652547A (en) * 1981-08-27 1987-03-24 Eli Lilly And Company Pharmaceutical formulations comprising human insulin and human proinsulin
US5053386A (en) * 1987-07-24 1991-10-01 Tung Ta C Orally administrable anti-metastatic lectin compositions and methods
US5340833A (en) * 1992-05-01 1994-08-23 Eisai Co., Ltd. Urokinase inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4791068A (en) * 1984-06-22 1988-12-13 Scripps Clinic And Research Foundation Diagnostic assay for inhibitor of tissue-type and urokinase-type plasminogen activators
DK319685A (da) * 1985-07-12 1987-01-13 Fonden Til Fremme Af Eksperime Monoklonale antistoffer, fremgangsmaade til frembringelse af antistofferne, hybridomaceller, der producerer antistofferne, og anvendelse af antistofferne
GB2246779B (en) * 1990-08-03 1994-08-17 Delta Biotechnology Ltd Tumour-associated protease inhibitors targeted to tumour cells

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4571421A (en) * 1979-11-05 1986-02-18 Genentech, Inc. Mammalian gene for microbial expression
US4652547A (en) * 1981-08-27 1987-03-24 Eli Lilly And Company Pharmaceutical formulations comprising human insulin and human proinsulin
WO1986006100A1 (en) * 1985-04-15 1986-10-23 Biotechnology Research Partners, Limited Human anti-inflammatory phospholipase inhibitor protein
US5053386A (en) * 1987-07-24 1991-10-01 Tung Ta C Orally administrable anti-metastatic lectin compositions and methods
US5340833A (en) * 1992-05-01 1994-08-23 Eisai Co., Ltd. Urokinase inhibitors

Non-Patent Citations (39)

* Cited by examiner, † Cited by third party
Title
Appella, E., et al., "The Receptor-binding Sequence of Urokinase," J. Biol. Chem., 262(10):4437-4440 (1987).*
Bajpai, A., et al., "Cryptic Urokinase Binding Sites on Human Foreskin Fibroblasts," Biochem. Biophys. Res. Commun., 133(2):475-482 (1985).*
Behrcndt, N., et al. (1991) ibid., 7842-47.*
Blasi, F., "Surface Receptors for Urokinase Plasminogen Activator," Fibrinolysis, 2:73-84 (1988).*
Blasi, F., et al., "The Receptor for Urokinase-Plasminogen Activator," J. Cell. Biochem., 32:179-186 (1986).*
Blasi, F., et al., "Urokinase-Type Plasminogen Activator: Proenzyme, Receptor, and Inhibitors," J. Cell. Biol., 104:801-804 (1987).*
Boyd, D., et al., "Determination of the Levels of Urokinase and Its Receptor in Human Colon Carcinoma Cell Lines," Cancer Research, 48:3112-3116 (Jun. 1, 1988).*
Boyd, D., et al., "Modulation of the urokinase receptor in human colon cell lines by N,N-dimethylformamide," Biochimica et Biophysica Acta, 970:96-100 (1988).*
Burtin, P., et al., "Receptor for Plasmin on Human Carcinoma Cells," J. Nat'l. Cancer Inst., 80(10):762-765 (Jul. 20, 1988).*
Cubeilis, M.V., et al., "Binding of Single-chain Prourokinase to the Urokinase Receptor of Human U937 Cells," J. Biol. Chem., 261(34):15819-15822, (Dec. 1986).*
Danø, K., et al., "Plasminogen Activators and Neoplasia," Tissue-Type Plasminogen Activator (t-PA): Physiological and Clinical Aspects, Kluft, ed., CRC Press, Boca Raton, pp. 19-46 (1988).*
Danø, K., et al., "Plasminogen Activators, Tissue Degradation, and Cancer," Advances in Cancer Research, 41:140-264, (1985).*
Ellis, V., et al., "Plasminogen Activation Initiated by a Single-Chain Urokinase-ytpe Plasminogen Activator," J. Biol. Chem., 264:2185-2188, (1988).*
Ellis, V., et al., "Role of human U937 monocytes in controlling single-chain urokinase-initiated plasminogen activation," Fibrinolysis, 2:112-114 (1988).*
Estreicher, A., et al., "Characterization of the Cellular Binding Site for the Urokinase-Type Plasminogen Activator," J. Biol. Chem., 264(2):1180-1189, (Jan. 1989).*
Hearing, V.J., et al., "Modulation of Metastatic Potential by Cell Surface Urokinase of Murine Melanoma Cells," Cancer Research, 48:1270-1278, (Mar., 1988).*
Hebert, C.A., et al., "Linkage of Extracellular Plasminogen Activator to the Fibroblast Cystoskeleton: Colocalization of Cell Surface Urokinase with Vinculin," J. Cell Biol., 106:1241-1247, (Apr., 1987).*
Kirchheimer, et al J. Immunol. vol. 141(12): pp. 4229-4234 1988.*
Mignatti, P., et al., "Tumor Invasion through the Human Amniotic Membrane: Requirement for a Proteinase Cascade," Cell, 47:487-498 (Nov. 1986).*
Miles, L.A., et al., "Receptor Mediated Binding of the Fibrinolytic Components, Plasminogen and Urokinase, to Peripheral Blood Cells," Thrombosis and Haemostasis, 58(3):936-942 (1987).*
Min, H. Y., et al. (1992) J. Immunol. 148 : 3636-42. *
Needham, G.K., et al., "Binding of urokinase to specific receptor sites on human breast cancer membranes," Br. J. Cancer, 55:13-16 (1987).*
Nelles, L., et al., "Characterization of Recombinant Human Single Chain Urokinase-type Plasminogen Activator Mutants Produced by Site-specific Mutagenesis of Lysine 158," J. Biol. Chem., 262(12):5682-89, (1987).*
Nielsen, L.S., et al., "A 55,000-60,000 Mr Receptor Protein for Urokinase-type Plasminogen Activator," J. Biol. Chem., 263(5):2358-2363, (Feb. 1988).*
Ossowski, L., "Plasminogen Activator Dependent Pathways in the Dissemination of Human Tumor Cells in the Chick Embryo," Cell, 52:321-328 (Feb. 1988).*
Ossowski, L., et al., "Antibodies to Plasminogen Activator Inhibit Human Tumor Metastasis," Cell, 35:611-619 (Dec. 1983).*
Pannell, R., et al., "Activation of Plasminogen by Single-Chain Urokinase or by Two-Chain Urokinase-A Demonstration That Single-Chain Urokinase Has A Low . . . ," Blood, 69(1):22-26 (Jan. 1987).*
Petersen, L.C., et al., "One-chain Urokinase-type Plasminogen Activator from Human Sarcoma Cells Is a Pro-enzyme with Little or No Intrinsic Activity," J. Biol. Chem., 263(23):11189-11195, (Aug. 1988).*
Picone, R., et al., "Regulation of Urokinase Receptors in Monocyte-like U937 Cells by Phorbol Ester PMA," J. Cell Biol., 1989.*
Ploug, M., et al. (1991) J. Biol. Chem. 266:1926-33.*
Plow, E.F., et al., "The Plasminogen System and Cell Surfaces: Evidence for Plasminogen and Urokinase Receptors on the Same Cell Type," J. Cell Biol., 103(6):2411-2420 (Dec. 1986).*
Pöllänen, J., et al., "Distinct Localizations of Urokinase-type Plasminogen Activator and Its Type 1 Inhibitor under Cultured Human Fibroblasts and Sarcoma Cells," J. Cell Biol., 104:1085-1096, (Apr. 1987).*
Pöllänen, J., et al., "Ultrastructural Localization of Plasma Membrane-associated Urokinase-type Plasminogen Activator at Focal Contacts," J. Cell Biol., 106:87-95, (Jan. 1988).*
Reich, R., et al., "Effects of Inhibitors of Plasminogen Activator, Serine Proteinases, and Collagenase IV on the Invasion of Basement Membranes by Metastatic Cells," Cancer Research, 48:3307-3312, (Jun. 1988).*
Saksela O., "Plasminogen activation and regulation of pericellular proteolysis," Biochimica et Biophysica Acta, 823:35-65, (1985).*
Skriver, L., et al., "Immunocytochemical Localization of Urokinase-type Plasminogen Activator in Lewis Lung Carcinoma," J. Cell Biol., 99:752-757, (Aug. 1984).*
Stoppelli, M.P., et al., "Autocrine Saturation of Pro-Urokinase Receptors on Human A431 Cells," Cell, 45:675-684, (Jun. 1986).*
Stoppelli, M.P., et al., "Differentiation-enhanced binding of the amino-terminal fragment of human urokinase plasminogen activator to a specific receptor on U937 monocytes," Proc. Natl. Acad. Sci. USA, 82:4939-43, 1985.*
Vassalli, J., et al., "A Cellular Binding Site for the Mr 55,000 Form of the Human Plasminogen Activator, Urokinase," J. Cell Biol., 100:86-92, (Jan. 1985).*

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090004176A1 (en) * 2001-11-02 2009-01-01 Paion Deutschland Gmbh Non-neurotoxic plasminogen activating factors for treating of stroke
US8119597B2 (en) 2001-11-02 2012-02-21 Paion Gmbh Non-neurotoxic plasminogen activating factors for treating of stroke
US8071091B2 (en) * 2001-11-02 2011-12-06 Paion Deutschland Gmbh Non-neurotoxic plasminogen activating factors for treating stroke
US20090263373A1 (en) * 2001-11-02 2009-10-22 Mariola Sohngen Non-neurotoxic plasminogen activating factors for treating of stroke
EP2390308A1 (de) 2002-02-25 2011-11-30 Vaxiion Therapeutics Inc Minizell-Zusammensetzungen und Verfahren
WO2005048822A3 (en) * 2003-11-18 2006-12-07 Attenuon Llc Antibodies and/or conjugates thereof which bind to the amino terminal fragment of urokinase, compositions and uses thereof
US7449196B2 (en) 2004-07-09 2008-11-11 Robert Sabin Anti tumor compositions and methods of use
US20060008535A1 (en) * 2004-07-09 2006-01-12 Robert Sabin Anti tumor compositions and methods of use
US20080280833A1 (en) * 2005-01-31 2008-11-13 Bernard Degryse Therapeutic Peptides Derived from Urokinase Plasminogen Activator Receptor
US20100075372A1 (en) * 2006-09-28 2010-03-25 Taka-Aki Sato Method for deparaffinization of paraffin-embedded specimen and method for analysis of paraffin-embedded specimen
US20080081051A1 (en) * 2006-09-28 2008-04-03 Robert Sabin Method of manufacturing anti-tumor and anti-viral compositions
US8105602B2 (en) 2006-12-08 2012-01-31 Tactic Pharma, Llc Urokinase-type plasminogen activator receptor epitope, monoclonal antibodies derived therefrom and methods of use thereof
US20080199476A1 (en) * 2006-12-08 2008-08-21 Graham Parry Urokinase-type plasminogen activator receptor epitope, monoclonal antibodies derived therefrom and methods of use thereof
GB2461410B (en) * 2006-12-22 2011-06-29 Hvidovre Hospital Method and tool for predicting cancer and other diseases
US20100098705A1 (en) * 2006-12-22 2010-04-22 Jesper Eugen-Olsen Method and tool for predicting cancer and other diseases
US9645157B2 (en) 2006-12-22 2017-05-09 Hvidovre Hospital Methods of selecting and treating subjects with low-grade inflammation and metabolic disorders
US8815519B2 (en) 2006-12-22 2014-08-26 Hvidovre Hospital Method for predicting cancer and other diseases
WO2008077958A2 (en) * 2006-12-22 2008-07-03 Hvidovre Hospital Soluble urokinase plasminogen activator receptor (supar) as marker for diseases
GB2461410A (en) * 2006-12-22 2010-01-06 Hvidovre Hospital Soluble urokinase plasminogen activator receptor (SUPAR) as marker for diseases
WO2008077958A3 (en) * 2006-12-22 2009-04-02 Hvidovre Hospital Soluble urokinase plasminogen activator receptor (supar) as marker for diseases
US8507646B2 (en) 2007-02-23 2013-08-13 Landing Biotech, Inc. Treating atherosclerosis
EP2126067A4 (de) * 2007-02-23 2010-05-19 Landing Biotech Inc Atherosklerose-behandlung
US20100143335A1 (en) * 2007-02-23 2010-06-10 Landing Biotech, Inc. Treating atherosclerosis
EP2126067A2 (de) * 2007-02-23 2009-12-02 Landing Biotech, Inc. Atherosklerose-behandlung
WO2008115870A2 (en) * 2007-03-16 2008-09-25 Mount Sinai School Of Medicine Induction and/or maintenance of tumor dormancy by disruption of urokinase plasminogen activator receptor-integrin interaction
WO2008115870A3 (en) * 2007-03-16 2009-01-08 Sinai School Medicine Induction and/or maintenance of tumor dormancy by disruption of urokinase plasminogen activator receptor-integrin interaction
US20100249184A1 (en) * 2007-03-16 2010-09-30 Mount Sinai School Of Medicine Induction and/or maintenance of tumor dormancy by disruption of urokinase plasminogen activator receptor-integrin interaction
US9333280B2 (en) 2009-02-25 2016-05-10 Teleflex Medical Incorporated Stabilized enzyme compositions
WO2015048619A3 (en) * 2013-09-30 2015-05-28 Teleflex Medical Incorporated Stabilized enzyme compositions
WO2020140101A1 (en) 2018-12-28 2020-07-02 Catalyst Biosciences, Inc. Modified urokinase-type plasminogen activator polypeptides and methods of use
US11613744B2 (en) 2018-12-28 2023-03-28 Vertex Pharmaceuticals Incorporated Modified urokinase-type plasminogen activator polypeptides and methods of use

Also Published As

Publication number Publication date
AU652917B2 (en) 1994-09-15
JP2002062295A (ja) 2002-02-28
EP1013285A3 (de) 2004-05-19
ATE197067T1 (de) 2000-11-15
WO1990012091A1 (en) 1990-10-18
AU683951B2 (en) 1997-11-27
DE467932T1 (de) 1993-01-14
JPH04506148A (ja) 1992-10-29
EP1013285A2 (de) 2000-06-28
CA2051676A1 (en) 1990-10-08
EP0467932B1 (de) 2000-10-18
EP0467932A1 (de) 1992-01-29
DE69033653D1 (de) 2000-11-23
DE69033653T2 (de) 2001-06-07
AU5528090A (en) 1990-11-05
DK0467932T3 (da) 2001-02-12
AU8046394A (en) 1995-04-13

Similar Documents

Publication Publication Date Title
US6248712B1 (en) Urokinase-type plasminogen activator receptor
US6113897A (en) Antibodies and their use
Wang The role and regulation of urokinase‐type plasminogen activator receptor gene expression in cancer invasion and metastasis
Krüger et al. Reduction of breast carcinoma tumor growth and lung colonization by overexpression of the soluble urokinase-type plasminogen activator receptor (CD87)
Rabbani et al. Urokinase receptor antibody can reduce tumor volume and detect the presence of occult tumor metastases in vivo
US5891664A (en) Vectors and methods for recombinant production of uPA-binding fragments of the human urokinase-type plasminogen receptor (uPAR)
EP0574391B1 (de) Antikörper gegen den urokinaserezeptor und ihre verwendung
Le Gall et al. Regulated cell surface pro-EGF ectodomain shedding is a zinc metalloprotease-dependent process
Zacharski et al. Pathways of coagulation activation in situ in rheumatoid synovial tissue
US20040213789A1 (en) Heparanase activity neutralizing anti-heparanase monoclonal antibody and other anti-heparanase antibodies
Radtke et al. Protein C inhibitor is expressed in tubular cells of human kidney.
Wojtukiewicz et al. Fibrin formation on vessel walls in hyperplastic and malignant prostate tissue
KR101154550B1 (ko) 폰 빌레브란트 인자 특이적 절단 효소에 대한 항체 및그것을 이용한 분석계
EP1064005B1 (de) Verwendung des urokinase plasminogen aktivator rezeptors als zielscheibe in der diagnose von micrometastasen
Jardi et al. Distinct patterns of urokinase receptor (uPAR) expression by leukemic cells and peripheral blood cells
EP0691350B1 (de) uPA-Bindungstelle auf 2+3 Domäne von uPAR und damit reagierende Antikörper
US20030138429A1 (en) Compositions and methods for inhibiting metastasis
Maréchal et al. EGF precursor mRNA and membrane-associated EGF precursor protein in rat exorbital lacrimal gland
US5422090A (en) Human PAI-2
Rappaport et al. Heparin Potentiates Endothelial Gell Growth Factor Stimulation of Plasminogen Activator Synthesis by Diploid Human Lung Fibroblasts
WILL et al. Expression of urokinase-type plasminogen-activator (uPA) and Its receptor (uPAR) in human ovarian-cancer cells and in-vitro invasion capacity
Combe Hepatic receptor (s) for serine protease-inhibitor complexes
CA2400497A1 (en) Methods and compositions for generating angiostatin
JP2003021632A (ja) ウロキナーゼレセプターに対する抗体とその用途
Sidenius Urokinase Receptor Cleavage and Shedding: Occurrence and Consequences

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: PAT HOLDER NO LONGER CLAIMS SMALL ENTITY STATUS, ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: STOL); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12